US20090093425A1 - Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups - Google Patents

Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups Download PDF

Info

Publication number
US20090093425A1
US20090093425A1 US11/776,317 US77631707A US2009093425A1 US 20090093425 A1 US20090093425 A1 US 20090093425A1 US 77631707 A US77631707 A US 77631707A US 2009093425 A1 US2009093425 A1 US 2009093425A1
Authority
US
United States
Prior art keywords
substituted
alkyl
alkenyl
aryl
alkoxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/776,317
Inventor
Steven F. Dowdy
Scott G. Petersen
Bryan R. Meade
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Howard Hughes Medical Institute
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of California filed Critical University of California
Priority to US11/776,317 priority Critical patent/US20090093425A1/en
Assigned to REGENTS OF THE UNIVERSITY OF CALIFORNIA, THE reassignment REGENTS OF THE UNIVERSITY OF CALIFORNIA, THE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DOWDY, STEVEN F., MEADE, BRYAN R., PETERSEN, SCOTT G.
Assigned to HOWARD HUGHES MEDICAL INSTITUTE, THE reassignment HOWARD HUGHES MEDICAL INSTITUTE, THE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PETERSEN, SCOTT G., DOWDY, STEVEN F.
Publication of US20090093425A1 publication Critical patent/US20090093425A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3513Protein; Peptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2330/00Production
    • C12N2330/30Production chemically synthesised

Definitions

  • This invention relates to compositions and methods for transducing cells.
  • Polyanionic oligomers do not readily diffuse across cell membranes.
  • cationic lipids when combined with anionic oligonucleotides are generally used to assist uptake.
  • this complex is generally toxic to cells, which means that both the exposure time and concentration of cationic lipid must be carefully controlled to insure transfection of viable cells.
  • Nucleic acid delivery to cells both in vitro and in vivo has been performed using various recombinant viral vectors and electroporation. Such techniques have sought to treat various diseases and disorders by knocking-out gene expression or providing genetic constructions for gene therapy.
  • RNA interference RNA interference
  • siRNAs RNA interference
  • siRNAs are macromolecules with no ability to enter cells. Indeed, siRNAs are 25 ⁇ in excess of Lipinski's “Rule of 5s” for cellular delivery of membrane diffusible molecules that generally limits size to less than 500 Da.
  • siRNAs do not enter cells, even at millimolar concentrations (Barquinero et al., Gene Ther. 11 Suppl 1, S3-9, 2004).
  • transfection reagents fail to achieve efficient delivery into many cell types, especially primary cells and hematopoietic cell lineages (T and B cells, macrophage).
  • lipofection reagents often result in varying degrees of cytotoxicity ranging from mild in tumor cells to high in primary cells.
  • Recent cell-directed targeting approaches using antibody fusions to DNA-condensing protamine (Song et al., Nat. Biotechnol. 23, 709-717, 2005) and siRNA fusions to receptor targeted RNA aptamers (McNamara et al., Nat. Biotechnol. 24, 1005-1015, 2006) offer the potential to delivery siRNAs into select cells.
  • the disclosure provides methods and compositions for delivering masked oligonucleotides or polynucleotides into living cells.
  • Reduced anionically charged, neutral and cationically charged oligonucleotides or polynucleotides either alone or conjugated to a transduction molecule traverse cell membranes better than anionically charged nucleic acid molecules.
  • Transiently protected oligonucleotides or polynucleotides comprising a protecting/charge neutralizing group can be synthesized by automated amidite methods. These compounds can enter the cytosol of living cells by endocytic or macropinocytic mechanisms.
  • the phosphotriester protecting/neutralizing group when exposed to the intracellular environment is designed to be removed by enzymatic activity or by passive intracellular methods (e.g., high intracellular concentrations of glutathione) to give phosphodiester oligonucleotides or polynucleotides capable of eliciting an RNAi response.
  • the disclosure provides oligonucleotide prodrugs useful as therapeutics, diagnostics and as tools for research.
  • the disclosure further provides a peptide RNA/DNA hybrid pro-drug where the phosphates that are normally found in a negatively charged state under physiological conditions along the backbone of an oligonucleotide or polynucleotide are transiently protected as a nonionic charge neutral phosphotriester.
  • the charge neutralizing phosphotriester group is formed by the addition of any number of different phosphate protecting groups (e.g., S-pivaloyl thioethanol (SPTE) group).
  • SPTE S-pivaloyl thioethanol
  • the phosphate backbone is protected with a biologically reversible R group that contains a positive charge, e.g., a guanidinium group via an enzymatically labile linker (RNB) to give net cationic character to the construct.
  • R group e.g., a guanidinium group via an enzymatically labile linker (RNB) to give net cationic character to the construct.
  • RNB enzymatically labile linker
  • the disclosure provides a nucleic acid construct comprising: a) an oligonucleotide or polynucleotide domain comprising a phosphodiester and/or phosphothioate protecting group that reduces the net anionic charge of the oligonucleotide or polynucleotide backbone; and b) a transduction domain comprising a membrane transport function operably linked to the oligonucleotide or polynucleotide domain.
  • the phosphodiester and/or phosphothioate protecting group has the general formula:
  • X is O, S or NR 1
  • R1 is H, methyl, ethyl, S-pivaloyl thioethanol, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • R 2 is selected from the group consisting of:
  • R 2 is alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic,
  • R 3 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 1 and A 2 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • R 4 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 3 and A 4 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • R 5 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 5 is a one to seven atom chain, or substituted one to seven atom chain,
  • X 1 and X 2 are each independently O, S or NR 7 , and R 7 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • R 6 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 6 and A 6 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • X 3 and X 4 are each independently O, S or NR 8 , and R 8 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • Q is selected from the group consisting of:
  • Q 1 is a basic group with a pKa greater than or equal to 10,
  • Q 2 is a basic group with a pKa greater than or equal to 10,
  • a 8 and A 9 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Q 3 is a basic group with a pKa greater than or equal to 10,
  • a 10 and A 11 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Q 4 is a basic group with a pKa greater than or equal to 10,
  • a 12 is a one to seven atom chain, or substituted one to seven atom chain,
  • X 5 and X 6 are each independently O, S or NR 9 , and R 9 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Q 5 is a basic group with a pKa greater than or equal to 10,
  • a 13 and A 14 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • X 7 and X 8 are each independently O, S or NR 10 , and R 10 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic.
  • the phosphodiester and/or phosphothioate protecting group is selected from the group consisting of MeO-, EtO-, iPrO,
  • the disclosure also provides a nucleic acid construct comprising: an oligonucleotide or polynucleotide comprising a phosphodiester and/or phosphothioate protecting group that reduces the net anionic charge of the oligonucleotide or polynucleotide backbone or provides a net cationic charge of the oligonucleotide or polynucleotide backbone.
  • the phosphodiester and/or phosphothioate protecting group has the general formula:
  • X is O, S or NR 1 , and R 1 is H, methyl, ethyl, S-pivaloyl thioethanol, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • R is selected from the group consisting of:
  • R 2 is alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic,
  • R 3 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 1 and A 2 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • R 4 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 3 and A 4 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • R 5 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 5 is a one to seven atom chain, or substituted one to seven atom chain,
  • X 1 and X 2 are each independently O, S or NR 7 , and R 7 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • R 6 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 6 and A 6 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • X 3 and X 4 are each independently O, S or NR 8 , and R 8 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • Q is selected from the group consisting of:
  • Q 1 is a basic group with a pKa greater than or equal to 10,
  • Q 2 is a basic group with a pKa greater than or equal to 10,
  • a 8 and A 9 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Q 3 is a basic group with a pKa greater than or equal to 10,
  • a 10 and A 11 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Q 4 is a basic group with a pKa greater than or equal to 10,
  • a 12 is a one to seven atom chain, or substituted one to seven atom chain,
  • X 5 and X 6 are each independently O, S or NR 9 , and R 9 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Q 5 is a basic group with a pKa greater than or equal to 10,
  • a 13 and A 14 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • X 7 and X 8 are each independently O, S or NR 10 , and R 10 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic.
  • the phosphodiester and/or phosphothioate protecting group is selected from the group consisting of MeO—, EtO—, iPrO,
  • the disclosure also provides pharmaceutical composition comprising the nucleic acid constructs described herein.
  • the disclosure describes a method comprising linking one or more protein transduction domain to a nucleic acid construct.
  • the one or more protein transduction domains comprise 2-5 protein transduction domains.
  • the disclosure also provides a method of generating a nucleic acid construct comprising: substantially purifying a protein transduction domain; synthesizing an oligonucleotide; charge neutralizing the anionic charge on the oligonucleotide with a phosphotriester group; and linking the oligonucleotide to one or more protein transduction domains.
  • Also provided are methods of transfecting a cell comprising contacting the cell with a nucleic acid construct of the disclosure.
  • the contacting can be in vivo or in vitro.
  • the nucleic acid construct can comprise an antisense molecule.
  • the disclosure also provides a method of treating a disease or disorder comprising administering a nucleic acid construct of the disclosure to a subject, wherein the oligonucleotide or polynucleotide comprises a therapeutic or diagnostic molecule.
  • FIG. 1 shows TAT and anionically charged oligonucleotide interactions.
  • TAT linked oligonucleotides lacking a phosphodiester and/or phosphothioate protecting group form complexes wherein the cationic charges are neutralized.
  • FIG. 2 depicts charge-neutralized oligonucleotides stimulating macropinocytosis.
  • FIG. 3 shows a general scheme of phosphotriester protection and charge neutralization during oligonucleotide uptake.
  • FIG. 4 show an embodiment of a PTD conjugated protected oligonucleotide and reversible deprotection.
  • FIG. 5 shows an embodiment a phosphodiester and/or phosphothioate protecting group formula.
  • FIG. 6 shows a nucleoside phosphoramidite synthesis route of the disclosure.
  • FIG. 7 depicts a reduction reaction for reversible charge neutralization.
  • FIG. 8 depicts an RNB phosphotriester protection and deprotection of anionic charged nucleic acids.
  • FIG. 9 shows chromatographic DTT reduction of a DTG protected uracil.
  • FIG. 10 shows redox sensitivity of DTG-Uridine phosphoramidite.
  • FIG. 11 shows dimer coupling reaction of DTG-Uridine phosphoramidite to di-TBS uridine.
  • FIG. 12 shows synthesis of poly-uridine oligos (SEQ ID NO:22) containing 2′ fluoride insertions with wildtype phosphodiester, methylphosphotriesters or SPTE-phosphotriesters. This figure shows the ability to synthesize RNA oligonucleotides containing phosphotriesters group with a 2′-Fluoro (F). Gel analysis performed under urea denaturing conditions followed by ethidium bromide staining (right panel).
  • FIG. 13A-D shows synthesis of double stranded siRNAs (SEQ ID NOs: 23-24) containing phosphotriester modifications.
  • A Structure of wild type phosphodiester and phosphotriester groups, methyl and SPTE are reversible, whereas the isopropyl group is not reversible and acts as a control.
  • B Sequence of eGFP siRNAs showing wild type Sense (S) strand (top line in panel) and Anti-Sense (AS) (also termed guide strand) (bottom line in panel) synthesized with six DNA thymidine insertions containing either wildtype, methyl, isopropyl or SPTE phosphotriesters as indicated.
  • AS Anti-Sense
  • C Single stranded RNA analysis of full length Anti-Sense (AS) oligonucleotides containing indicated phosphotriester modifications. Gel analysis performed under urea denaturing conditions followed by ethidium bromide staining.
  • D Double stranded RNA analysis of synthetic duplexes containing phosphotriester modifications.
  • Sense (S) strand (top line) is wild type RNA.
  • Anti-Sense (AS) strand contains 6 ⁇ wild type DNA Thymidines (AS-6T), 6 ⁇ methyl-phosphotriester Thymidines (AS-Met6T), 6 ⁇ isopropyl-phosphotriester Thymidines (AS-Iso6T), 6 ⁇ SPTE-phosphotriester Thymidines (AS-SPTE6T), as indicated.
  • Gel analysis performed under non-denaturing conditions followed by ethidium bromide staining. This data shows the ability to make stable, full length siRNAs containing phosphotriester modifications.
  • FIG. 14A-B shows analysis of GFP reporter RNA Interference (RNAi) response.
  • A Flow cytometry (FACS) of reporter H1299 cells stably expressing eGFP following treatment with indicated siRNA duplexes. Cells were transfected with 50 nM indicated siRNAs plus Lipofectamine 2000 for 4 hr then analyzed for GFP expression by FACS at 24 hr. Untreated control cells show high level of GFP expression. Transfection of irreversible S/AS-Iso6T RNA showed no change in GFP expression, indicating a blocking or failure to induce an RNAi response by the continued presence of the 6 ⁇ isopropyl phosphotriester groups.
  • FIG. 15A-B shows PTD mediated delivery of charge neutralized, dsDNA.
  • FACS flow cytometry
  • dsDNA is only taken up when conjugated to either 2 ⁇ or 3 ⁇ PTD peptides via a disulfide bond (2 ⁇ /3 ⁇ PTD-S—S-15N/15N-Cy3), whereas control of peptide added to dsDNA, but NOT conjugated (2 ⁇ /3 ⁇ PTD+15N/15N-Cy3), is not taken up by cells.
  • B Dose curve graph of PTD-dsDNA-Cy3 oligo conjugates from above after 4 hour treatment of H1299 cells by FACS as indicated.
  • the ability to deliver certain bioactive agents to the interior of cells is problematical due to the bioavailability restriction imposed by the cell membrane.
  • the plasma membrane of the cell forms a barrier that restricts the intracellular uptake of molecules to those which are sufficiently non-polar and smaller than approximately 500 daltons in size.
  • Previous efforts to enhance the cellular internalization of proteins have focused on fusing proteins with receptor ligands (Ng et al., Proc. Natl. Acad. Sci. USA, 99:10706-11, 2002) or by packaging them into caged liposomal carriers (Abu-Amer et al., J. Biol. Chem. 276:30499-503, 2001).
  • these techniques often result in poor cellular uptake and intracellular sequestration into the endocytic pathway.
  • the disclosure provides methods and compositions to facilitate and improve cellular uptake of nucleic acid molecules by “protecting” the charge associated with an oligonucleotide or polynucleotide.
  • RNA aptamers have great potential to bind to, sequester and inhibit proteins, but at >10,000 Daltons and highly charged, they have no or limited ability to enter cells on their own.
  • the methods and compositions of the disclosure allow for intracellular delivery of RNA aptamers, siRNA and DNA vectors.
  • sequence specific oligonucleotides or polynucleotides to selectively treat human diseases can more effectively be delivering useful oligonucleotides and polynucleotides, including siRNAs, RNA aptamers, and DNA vectors to subjects and to cells.
  • the disclosure overcomes size and charge limitations that make such RNAi constructs difficult to deliver or undeliverable.
  • a construct comprising a phosphodiester and/or phosphothioate protecting group can deliver nucleic acids into a cell in vitro and in vivo.
  • the disclosure provides nucleic acid constructs comprising phosphodiester and/or phosphothioate protecting groups.
  • the construct can further include compositions useful in cellular transduction and cellular modulation.
  • Such compositions can include transduction moiety domains comprising a membrane transport function and may further comprise a nucleic acid binding domain sufficient to reversibly neutralize anionic charges on nucleic acids.
  • charge neutralization of anionic nucleic acid e.g., an RNA molecule
  • anionic nucleic acid e.g., an RNA molecule
  • a phosphodiester and/or phosphothioate protecting group(s) promotes uptake.
  • the charge neutralized anionic nucleic acid is linked to a PTD the charge neutralization of the anionic charged nucleic acid frees the cationic PTD to traverse the membrane as well as prevents aggregation of the conjugate.
  • the exposed free cationic charge of the PTD can then effectively interact with a cell surface, induce macropinocytosis and escape from the macropinosome into the cytoplasm.
  • the phosphodiester and/or phosphothioate protecting group(s) can be removed by intracellular processes, such as reduction of a disulfide linkage or ester hydrolysis, allowing for removal from the construct in the cytoplasm.
  • a nucleic acid construct that includes, for example, dsRNA can then be hydrolyzed by Dicer, an RNAse III-like ribonuclease, thereby releasing siRNA that silences a target gene.
  • nucleic acid can effectively facilitate cell transduction.
  • Any nucleic acid regardless of sequence composition, can be modified by phosphodiester and/or phosphothioate protecting group(s).
  • the disclosure provides oligonucleotides or polynucleotides having, in some embodiments, one or more bioreversible protecting groups that contribute to chemical and biophysical properties that enhance cellular membrane penetration and resistance to exo- and endonuclease degradation.
  • the disclosure further provided amidite reagents for the synthesis of the bioreversible protected oligonucleotides or polynucleotides. Moreover, these protecting groups are stable during the synthetic processes.
  • the oligonucleotides or polynucleotides of the disclosure having one or more bioreversible protecting groups are sometimes referred to as pro-oligonucleotides or pro-polynucleotides.
  • the pro-oligonucleotides are capable of improved cellular lipid bilayers penetrating potential as well as resistance to exo- and endonuclease degradation in vivo.
  • the bioreversible protecting groups are removed in the cell cytosol by reducing conditions, enzymatic activity (e.g., endogenous carboxyesterases) and the like to yield biologically active oligonucleotide compounds that are capable of hybridizing to and/or having an affinity for specific endogenous nucleic acids.
  • enzymatic activity e.g., endogenous carboxyesterases
  • the phosphodiester and/or phosphothioate protecting groups can be used with antisense oligonucleotides of synthetic DNA or RNA or mixed molecules of complementary sequences to a target sequence belonging to a gene or to an RNA messenger whose expression they are specifically designed to block or down-regulate.
  • the antisense oligonucleotides may be directed against a target messenger RNA sequence or, alternatively against a target DNA sequence, and hybridize to the nucleic acid to which they are complementary. Accordingly, these molecules effectively block or down-regulate gene expression.
  • Protected oligonucleotides or polynucleotides may also be directed against certain bicatenary DNA regions (homopurine/homopyrimidine sequences or sequences rich in purines/pyrimidines) and thus form triple helices.
  • the formation of a triple helix, at a particular sequence, can block the interaction of protein factors which regulate or otherwise control gene expression and/or may facilitate irreversible damage to be introduced to a specific nucleic acid site if the resulting oligonucleotide is made to possess a reactive functional group.
  • nucleic acid constructs and methods of producing such constructs, that can be used for facilitating the delivery of oligonucleotides or polynucleotides in to cells.
  • a nucleic acid construct includes one or more phosphodiester and/or phosphothioate protecting group(s) to neutralize the phosphodiester anionic charge associated with a nucleic acid, such as RNA and/or DNA.
  • the protecting group can be removed from the construct by intracellular processes that include disulfide linkage reduction, ester hydrolysis or other enzyme-mediated processes.
  • the nucleic acid construct comprising one or more phosphodiester and/or phosphothioate protecting group further comprises one or more transduction domains such as a protein transduction domain (PTD).
  • a PTD can be conjugated directly to an oligonucleotide (e.g., an RNA or DNA) comprising the nucleic acid construct, such as at the 5′ and/or 3′ end via a free thiol group.
  • a PTD can be linked to the construct by a biologically sensitive and reversible manner, such as a disulfide linkage. This approach can be applied to any oligonucleotide or polynucleotide length and will allow for delivery of RNA (e.g., siRNA, RNA apatmer) or DNA into cells.
  • RNA e.g., siRNA, RNA apatmer
  • a nucleic acid construct can include a basic group, such as guanidium group (similar to the head group arginine, an active component of the PTD), linked to the reversible protecting group and thereby limit the need for the PTD.
  • a basic group such as guanidium group (similar to the head group arginine, an active component of the PTD), linked to the reversible protecting group and thereby limit the need for the PTD.
  • nucleic acid constructs synthesized to include phosphodiester and/or phosphothioate protecting group(s) for the delivery of nucleic acid sequences across a cell membrane.
  • the construct can also include, for example, one or more transduction domains and/or a protecting group that contains a basic group.
  • An isolated nucleic acid construct refers to an oligonucleotide or polynucleotide associated with a molecule or compound comprising a phosphodiester and/or phosphothioate protecting group.
  • a nucleic acid construct includes, but is not limited to, an oligonucleotide or polynucleotide associated with an anionic charge reducing group(s) or molecule(s), by hydrogen bonding, charge association, covalent bonding and the like, to promote uptake by a cell.
  • anionic charge reducing molecules that can be associated with an oligonucleotide or polynucleotide in the nucleic acid constructs of the disclosure include fusion polypeptides or peptides, chemical moieties that reduce the net anionic charge of an oligonucleotide or polynucleotide and combinations thereof.
  • nucleic acid constructs include selective treatment of cancer, viral infection, genetic diseases, nucleic acid delivery for research and the like.
  • polynucleotide(s) and oligonucleotide(s) generally refers to any polyribonucleotide or polydeoxyribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • an oligonucleotide as used herein refers to, among others, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions.
  • a oligonucleotide can comprise an siRNA, an antisense molecule, a ribozyme and the like.
  • a polynucleotide or oligonucleotides also includes triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • the strands in such regions may be from the same molecule or from different molecules.
  • the regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules.
  • a polynucleotide or oligonucleotide includes DNAs or RNAs as described above that contain one or more modified bases.
  • DNAs or RNAs with backbones comprising unusual bases, such as inosine, or modified bases, such as tritylated bases are polynucleotides or oligonucleotides as the term is used herein.
  • nucleic acid construct comprising an oligonucleotide with reduced anionic charged and including a phosphodiester and/or phosphothioate protecting group or charge neutralization group.
  • the construct can further include transduction domains and/or nucleic acid binding domains.
  • RNA binding domain e.g., TAT-DRBD
  • TAT-DRBD double stranded RNA binding domain
  • an anionic charge neutralizing molecule or group refers to a molecule or chemical group that can reduce the overall net anionic charge of an oligonucleotide or polynucleotide to which it is associated.
  • Phosphodiester and/or phosphothioate protecting groups as described herein are anionic charge neutralizing groups.
  • the phosphodiester and/or phosphothioate protecting groups can be reversible or irreversible.
  • One or more anionic charge neutralizing molecules or groups can be associated with an oligonucleotide or polynucleotide wherein each independently contributes to a reduction or the anionic charge and or increase in cationic charge of the construct.
  • one or more phosphodiester and/or phosphothioate protecting groups can be associated with an oligonucleotide and the “protected oligonucleotide” associated with one or more cationic transduction domains (e.g., PTDs), such that the overall net anionic charge of the construct is reduced or the overall net charge of the construct is neutral or the overall net charge of the construct is cationic relative to the oligonucleotide without the phosphodiester and/or phosphothioate protecting group and/or PTD.
  • PTDs cationic transduction domains
  • the disclosure provides phosphodiester and/or phosphothioate protecting groups and oligonucleotides or polynucleotides comprising such phosphodiester and/or phosphothioate protecting groups.
  • the phosphodiester and/or phosphothioate protecting group has the general formula:
  • X is O, S or NR 1
  • R1 is H, methyl, ethyl, S-pivaloyl thioethanol, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • R is selected from the group consisting of:
  • R 2 is alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic,
  • R 3 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 1 and A 2 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • R 4 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 3 and A 4 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • R 5 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 5 is a one to seven atom chain, or substituted one to seven atom chain,
  • X 1 and X 2 are each independently O, S or NR 7 , and R 7 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • R 6 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • a 6 and A 6 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • X 3 and X 4 are each independently O, S or NR 8 , and R 8 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • Q is selected from the group consisting of:
  • Q 1 is a basic group with a pKa greater than or equal to 10,
  • Q 2 is a basic group with a pKa greater than or equal to 10,
  • a 8 and A 9 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Q 3 is a basic group with a pKa greater than or equal to 10,
  • a 10 and A 11 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Q 4 is a basic group with a pKa greater than or equal to 10,
  • a 12 is a one to seven atom chain, or substituted one to seven atom chain,
  • X 5 and X 6 are each independently O, S or NR 9 , and R 9 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Q 5 is a basic group with a pKa greater than or equal to 10,
  • a 13 and A 14 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • X 7 and X 8 are each independently O, S or NR 10 , and R 10 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic.
  • the phosphodiester and/or phosphothioate protecting group is selected from the group consisting of MeO—, EtO—, iPrO,
  • the phosphodiester and/or phosphothioate protecting group comprises a structure selected from the group consisting of:
  • the RNB protecting group comprises a structure selected from the group consisting of:
  • Alkyl groups include straight-chain, branched and cyclic alkyl groups. Alkyl groups include those having from 1 to 20 carbon atoms. Alkyl groups include small alkyl groups having 1 to 3 carbon atoms. Alkyl groups include medium length alkyl groups having from 4-10 carbon atoms. Alkyl groups include long alkyl groups having more than 10 carbon atoms, particularly those having 10-20 carbon atoms. Cyclic alkyl groups include those having one or more rings. Cyclic alkyl groups include those having a 3-, 4-, 5-, 6-, 7-, 8-, 9- or 10-member carbon ring and particularly those having a 3-, 4-, 5-, 6-, or 7-member ring.
  • the carbon rings in cyclic alkyl groups can also carry alkyl groups.
  • Cyclic alkyl groups can include bicyclic and tricyclic alkyl groups.
  • Alkyl groups optionally include substituted alkyl groups.
  • Substituted alkyl groups include among others those which are substituted with aryl groups, which in turn can be optionally substituted.
  • alkyl groups include methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, n-butyl, s-butyl, t-butyl, cyclobutyl, n-pentyl, branched-pentyl, cyclopentyl, n-hexyl, branched hexyl, and cyclohexyl groups, all of which are optionally substituted.
  • Alkenyl groups include straight-chain, branched and cyclic alkenyl groups. Alkenyl groups include those having 1, 2 or more double bonds and those in which two or more of the double bonds are conjugated double bonds. Alkenyl groups include those having from 2 to 20 carbon atoms. Alkenyl groups include small alkyl groups having 2 to 3 carbon atoms. Alkenyl groups include medium length alkenyl groups having from 4-10 carbon atoms. Alkenyl groups include long alkenyl groups having more than 10 carbon atoms, particularly those having 10-20 carbon atoms. Cyclic alkenyl groups include those having one or more rings.
  • Cyclic alkenyl groups include those in which a double bond is in the ring or in an alkenyl group attached to a ring. Cyclic alkenyl groups include those having a 3-, 4-, 5-, 6-, 7-, 8-, 9- or 10-member carbon ring and particularly those having a 3-, 4-, 5-, 6- or 7-member ring. The carbon rings in cyclic alkenyl groups can also carry alkyl groups. Cyclic alkenyl groups can include bicyclic and tricyclic alkyl groups. Alkenyl groups are optionally substituted. Substituted alkenyl groups include among others those which are substituted with alkyl or aryl groups, which groups in turn can be optionally substituted.
  • alkenyl groups include ethenyl, prop-1-enyl, prop-2-enyl, cycloprop-1-enyl, but-1-enyl, but-2-enyl, cyclobut-1-enyl, cyclobut-2-enyl, pent-1-enyl, pent-2-enyl, branched pentenyl, cyclopent-1-enyl, hex-1-enyl, branched hexenyl, cyclohexenyl, all of which are optionally substituted.
  • Aryl groups include groups having one or more 5- or 6-member aromatic or heteroaromatic rings.
  • Aryl groups can contain one or more fused aromatic rings.
  • Heteroaromatic rings can include one or more N, O, or S atoms in the ring.
  • Heteroaromatic rings can include those with one, two or three N, those with one or two 0, and those with one or two S.
  • Aryl groups are optionally substituted.
  • Substituted aryl groups include among others those which are substituted with alkyl or alkenyl groups, which groups in turn can be optionally substituted.
  • Specific aryl groups include phenyl groups, biphenyl groups, pyridinyl groups, and naphthyl groups, all of which are optionally substituted.
  • Arylalkyl groups are alkyl groups substituted with one or more aryl groups wherein the alkyl groups optionally carry additional substituents and the aryl groups are optionally substituted.
  • Specific alkylaryl groups are phenyl-substituted alkyl groups, e.g., phenylmethyl groups.
  • Alkylaryl groups are aryl groups substituted with one or more alkyl groups wherein the alkyl groups optionally carry additional substituents and the aryl groups are optionally substituted.
  • Specific alkylaryl groups are alkyl-substituted phenyl groups such as methylphenyl.
  • the rings that may be formed from two or more of R1-R5 together can be optionally substituted cycloalkyl groups, optionally substituted cycloalkenyl groups or aromatic groups.
  • the rings may contain 3, 4, 5, 6, 7 or more carbons.
  • the rings may be heteroaromatic in which one, two or three carbons in the aromatic ring are replaced with N, O or S.
  • the rings may be heteroalkyl or heteroalkenyl, in which one or more CH2 groups in the ring are replaced with O, N, NH, or S.
  • Optional substitution of any alkyl, alkenyl and aryl groups includes substitution with one or more of the following substituents: halogens, —CN, —COOR, —OR, —COR, —OCOOR, —CON(R) 2, —OCON(R) 2, —N(R) 2, —NO2, —SR, —SO2R, —SO2N(R) 2 or —SOR groups.
  • Optional substitution of alkyl groups includes substitution with one or more alkenyl groups, aryl groups or both, wherein the alkenyl groups or aryl groups are optionally substituted.
  • Optional substitution of alkenyl groups includes substitution with one or more alkyl groups, aryl groups, or both, wherein the alkyl groups or aryl groups are optionally substituted.
  • Optional substitution of aryl groups includes substitution of the aryl ring with one or more alkyl groups, alkenyl groups, or both, wherein the alkyl groups or alkenyl groups are optionally substituted.
  • Optional substituents for alkyl, alkenyl and aryl groups include among others:
  • R is a hydrogen or an alkyl group or an aryl group and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl groups all of which are optionally substituted;
  • R is a hydrogen, or an alkyl group or an aryl groups and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted;
  • R independently of each other R is a hydrogen or an alkyl group or an aryl group and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted; R and R can form a ring which may contain one or more double bonds;
  • R independently of each other R, is a hydrogen or an alkyl group or an aryl group and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted; R and R can form a ring which may contain one or more double bonds;
  • each R independently of each other R, is a hydrogen, or an alkyl group, acyl group or an aryl group and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl or acetyl groups all of which are optionally substituted; or R and R can form a ring which may contain one or more double bonds.
  • R is an alkyl group or an aryl groups and more specifically where R is methyl, ethyl, propyl, butyl, phenyl groups all of which are optionally substituted; for —-SR, R can be hydrogen;
  • R is a hydrogen, an alkyl group, or an aryl group and R and R can form a ring;
  • R can be an acyl yielding—OCOR* where R* is a hydrogen or an alkyl group or an aryl group and more specifically where R* is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted.
  • Specific substituted alkyl groups include haloalkyl groups, particularly trihalomethyl groups and specifically trifluoromethyl groups.
  • Specific substituted aryl groups include mono-, di-, tri, tetra- and pentahalo-substituted phenyl groups; mono-, di-, tri-, tetra-, penta-, hexa-, and hepta-halo-substituted naphthalene groups; 3- or 4-halo-substituted phenyl groups, 3- or 4-alkyl-substituted phenyl groups, 3- or 4-alkoxy-substituted phenyl groups, 3- or 4-RCO-substituted phenyl, 5- or 6-halo-substituted naphthalene groups.
  • substituted aryl groups include acetylphenyl groups, particularly 4-acetylphenyl groups; fluorophenyl groups, particularly 3-fluorophenyl and 4-fluorophenyl groups; chlorophenyl groups, particularly 3-chlorophenyl and 4-chlorophenyl groups; methylphenyl groups, particularly 4-methylphenyl groups, and methoxyphenyl groups, particularly 4-methoxyphenyl groups.
  • oligonucleotide or polynucleotide is linked to a PTD
  • charge neutralization of the anionically charged oligonucleotide or polynucleotide frees the cationically charged PTD to productively interact with the cell surface and also prevents aggregation of the conjugate.
  • the exposed cationic charged PTD interacts with the cell surface and induces macropinocytosis.
  • the oligonucleotide is released into the cytoplasm.
  • the protecting group can be cleaved off by cellular processes, such as a reducing enzyme, oxidizing enzyme, reducing agent, oxidizing agent or esterase, unprotecting the oligonucleotide or polynucleotide allowing the nucleic acid to revert to its natural configuration.
  • cellular processes such as a reducing enzyme, oxidizing enzyme, reducing agent, oxidizing agent or esterase, unprotecting the oligonucleotide or polynucleotide allowing the nucleic acid to revert to its natural configuration.
  • a nucleic acid domain used interchangeably with oligonucleotide or polynucleotide domain, can be any oligonucleotide or polynucleotide (e.g., a ribozyme, antisense molecule, polynucleotide, oligonucleotide and the like).
  • Oligonucleotides or polynucleotides generally contain phosphodiester bonds, although in some cases, nucleic acid analogs are included that may have alternate backbones, comprising, e.g., phosphoramidate, phosphorothioate, phosphorodithioate, or O-methylphophoroamidite linkages (see Eckstein, Oligonucleotides and Analogues: A Practical Approach, Oxford University Press); and peptide nucleic acid backbones and linkages.
  • Other analog nucleic acids include those with positive backbones; non-ionic backbones, and non-ribose backbones, including those described in U.S. Pat. Nos.
  • nucleic acids containing one or more carbocyclic sugars are also included within one definition of nucleic acids. Modifications of the ribose-phosphate backbone may be done for a variety of reasons, e.g. to increase the stability and half-life of such molecules in physiological environments. Mixtures of naturally occurring nucleic acids and analogs are encompassed by the term oligonucleotide and polynucleotide; alternatively, mixtures of different nucleic acid analogs, and mixtures of naturally occurring nucleic acids and analogs can be made. Furthermore, hybrids of RNN, RNB, DNA, and RNA can be used. dsDNA, ssDNA, dsRNA, siRNA are encompassed by the term oligonucleotide and polynucleotide.
  • a polynucleotide refers to a polymeric compound made up of any number of covalently bonded nucleotide monomers, including nucleic acid molecules such as DNA and RNA molecules, including single- double- and triple-stranded such molecules, and is expressly intended to embrace that group of polynucleotides commonly referred to as “oligonucleotides”, which are typically distinguished as having a relatively small number (no more than about 30, e.g., about 5-10, 10-20 or 20-30) of nucleotide constituents.
  • RNA is an abbreviation for “short interfering RNA”, also sometimes known as “small interfering RNA” or “silencing RNA”, and refers to a class of about 19-25 nucleotide-long double-stranded ribonucleic acid molecules that in eukaryotes are involved in the RNA interference (RNAi) pathway that results in post-transcriptional, sequence-specific gene silencing.
  • RNAi RNA interference
  • dsRNA is an abbreviation for “double-stranded RNA” and as used herein refers to a ribonucleic acid molecule having two complementary RNA strands and which stands distinct from siRNA in being at least about 26 nucleotides in length, and more typically is at least about 50 to about 100 nucleotides in length.
  • the nucleic acid may be DNA, both genomic and cDNA, RNA or a hybrid, where the nucleic acid may contain combinations of deoxyribo- and ribo-nucleotides, and combinations of bases, including uracil, adenine, thymine, cytosine, guanine, inosine, xanthine hypoxanthine, isocytosine, isoguanine, etc.
  • nucleoside includes nucleotides and nucleoside and nucleotide analogs, and modified nucleosides such as amino modified nucleosides.
  • nucleoside includes non-naturally occurring analog structures. Thus, e.g. the individual units of a peptide nucleic acid, each containing a base, are referred to herein as a nucleoside.
  • nucleic acid domain of a nucleic acid construct described herein is not limited by any particular sequence. Any number of oligonucleotide or polynucleotides useful for diagnostics, therapeutics and research can be used in the methods and compositions of the disclosure. Various sources of oligonucleotides and polynucleotides are available to one of skill in the art.
  • fragments of a genome may be isolated and the isolated polynucleotides modified in accordance with the disclosure to reduce the overall net anionic charge using phosphodiester and/or phosphothioate protecting groups or may be used as a source for extension of the oligonucleotide or polynucleotide using, for example, nucleic acid synthesis techniques known in the art.
  • Nucleic acid synthesizers are commercially available and their use is generally understood by persons of ordinary skill in the art as being effective in generating nearly any oligonucleotide of reasonable length which may be desired.
  • phosphoramidite chemistry useful 5′OH sugar blocking groups abbreviated to DMT in the figures, are trityl, momomethoxytrityl, dimethoxytrityl and trimethoxytrityl, especially dimethoxytrityl (DMTr).
  • useful phosphite activating groups i.e., NR 2
  • One approach includes the use of the di-isopropylamino activating group.
  • nucleoside units can be can be activated as amidites of the disclosure and incorporated in to the oligonucleotides or polynucleotides of the disclosure.
  • These include deoxy nucleotides, i.e., wherein W in the above structures is H, ribonucleotides, in some embodiment W is F, i.e., wherein W is OH in the above structures, 2′-alkoxy nucleotides, i.e., wherein W is O-alkyl in the above structures, or substituted 2′-O-alkyl nucleotides, i.e., wherein W is substituted —O-alkyl in the above structures.
  • 2′-O-alkyl nucleotides are described in U.S. Pat. No. 5,466,786, herein incorporated by reference.
  • a particularly useful substituted 2′-O-alkyl group, the methoxyethoxy group, is described by Martin, P., Helv. Chim. Acta, 1995, 78, 486-504, also herein incorporated by reference.
  • Oligonucleotides can be synthesized by a Mermade ⁇ 6 solid phase automated oligonucleotide synthesizer or any commonly available automated oligonucleotide synthesizer. Triester, phosphoramidite, or hydrogen phosphonate coupling chemistries described in, for example, M. Caruthers, Oligonucleotides: Antisense Inhibitors of Gene Expression., pp. 7-24, J. S. Cohen, ed. (CRC Press, Inc. Boca Raton, Fla., 1989) or Oligonucleotide synthesis, a practical approach, Ed. M. J. Gait, IRL Press, 1984; “Oligonucleotides and Analogues, A Practical Approach”, Ed.
  • the Beaucage reagent as described in, for example, Journal of American Chemical Society, 1990, 112, 1253-1255, or elemental sulfur, as described in Beaucage et al., Tetrahedron Letters, 1981, 22, 1859-1862, is used with phosphoramidite or hydrogen phosphonate chemistries to provide substituted phosphorothioate oligonucleotides.
  • the reagents comprising the protecting groups recited herein can be used in numerous applications where protection is desired.
  • nucleoside and nucleotide analogs are also contemplated by this disclosure to provide oligonucleotide or oligonucleoside analogs bearing the protecting groups disclosed herein.
  • nucleoside, nucleotide, deoxynucleoside and deoxynucleotide generally include analogs such as those described herein.
  • analogs are those molecules having some structural features in common with a naturally occurring nucleoside or nucleotide such that when incorporated into an oligonucleotide or oligonucleoside sequence, they allow hybridization with a naturally occurring oligonucleotide sequence in solution.
  • these analogs are derived from naturally occurring nucleosides and nucleotides by replacing and/or modifying the base, the ribose or the phosphodiester moiety. The changes can be tailor made to stabilize or destabilize hybrid formation or enhance the specificity of hybridization with a complementary nucleic acid sequence as desired.
  • structural groups are optionally added to the ribose or base of a nucleoside for incorporation into an oligonucleotide, such as a methyl, propyl or allyl group at the 2′-0 position on the ribose, or a fluoro group which substitutes for the 2′-O group, or a bromo group on the ribonucleoside base.
  • a methyl, propyl or allyl group at the 2′-0 position on the ribose or a fluoro group which substitutes for the 2′-O group, or a bromo group on the ribonucleoside base.
  • amidite reagents are commercially available, including 2′-deoxy amidites, 2′-O-methyl amidites and 2′-O-hydroxyl amidites. Any other means for such synthesis may also be employed.
  • oligonucleotides The actual synthesis of the oligonucleotides is well within the talents of those skilled in the art. It is also well known to use similar techniques to prepare other oligonucleotides such as the phosphorothioates, methyl phosphonates and alkylated derivatives. It is also well known to use similar techniques and commercially available modified amidites and controlled-pore glass (CPG) products such as biotin, Cy3, fluorescein, acridine or psoralen-modified amidites and/or CPG (available from Glen Research, Sterling Va.) to synthesize fluorescently labeled, biotinylated or other conjugated oligonucleotides.
  • CPG controlled-pore glass
  • phosphotriester neutralizing/protecting groups described herein are useful for neutralizing the anionic charge of a nucleic acid domain
  • additional cationically charged moieties linked to a protected nucleic acid domain can be used to further facilitate uptake of oligonucleotide and polynucleotides.
  • Macropinocytosis is a nonselective form of endocytosis that all cells perform.
  • herpes simplex virus structural protein VP22 Elliott and O'Hare, Cell 88:223-33, 1997)
  • the HIV-1 transcriptional activator TAT protein Green and Loewenstein, Cell 55:1179-1188, 1988; Frankel and Pabo, Cell 55:1189-1193, 1988
  • the cationic N-terminal domain of prion proteins Not only can these proteins pass through the plasma membrane but the attachment of other proteins, such as the enzyme ⁇ -galactosidase, was sufficient to stimulate the cellular uptake of these complexes.
  • Such chimeric proteins are present in a biologically active form within the cytoplasm and nucleus.
  • a protein transduction domain PTD
  • a heterologous molecule e.g., a polynucleotide, small molecule, or protein
  • PTDs are typically cationic in nature. These cationic protein transduction domains track into lipid raft endosomes carrying with them their linked cargo and release their cargo into the cytoplasm by disruption of the endosomal vesicle.
  • Examples of PTDs include AntHD, TAT, VP22, cationic prion protein domains, poly-Arg, AGRKKRRQRRR (SEQ ID NO:15), YARKARRQARR (SEQ ID NO:16), YARAAARQARA (SEQ ID NO:17), YARAARRAARR (SEQ ID NO:18), YARAARRAARA (SEQ ID NO:19), YARRRRRRRRR (SEQ ID NO:20), YAAARRRRRRR (SEQ ID NO:21) and functional fragments and variants thereof.
  • the disclosure provides, in one aspect, methods and compositions that combine the use of PTDs such as TAT and poly-Arg, with a charge neutralized nucleic acids.
  • charge neutralized is meant that the anionic charge of the nucleic acid (e.g., oligonucleotide or polynucleotide) is reduced, neutralized or more cationic than the same nucleic acid in the absence of a phosphodiester and/or phosphothioate protecting group or a phosphodiester and/or phosphothioate protecting group and a binding domain capable of neutralizing the anionic charge on a nucleic acid (i.e., the “cargo”) domain.
  • the nucleic acid e.g., oligonucleotide or polynucleotide
  • the transduction domain of a nucleic acid construct of the disclosure can be nearly any synthetic or naturally-occurring amino acid sequence that can transduce or assist in the transduction of the fusion molecule.
  • the transduction domain is cationically charged.
  • transduction can be achieved in accord with the disclosure by use of a nucleic acid construct including phosphodiester and/or phosphothioate protecting groups and a protein sequence such as an HIV TAT protein or fragment thereof that is linked at the N-terminal or C-terminal end to an oligonucleotide or polynucleotide comprising a phosphodiester and/or phosphothioate protecting group.
  • the nucleic acid may comprise a phosphodiester and/or phosphothioate protecting group and may also comprise a nucleic acid binding domain (e.g., a DRBD).
  • the transducing protein domain for example, can be the Antennapedia homeodomain or the HSV VP22 sequence, the N-terminal fragment of a prion protein or suitable transducing fragments thereof such as those known in the art.
  • PTDs will be capable of transducing at least about 20%, 25%, 50%, 75%, 80%, 90%, 95%, 98% 99% or 100% of the cells.
  • Transduction efficiency typically expressed as the percentage of transduced cells, can be determined by several conventional methods.
  • PTDs will manifest cell entry and exit rates (sometimes referred to as k 1 and k 2 , respectively) that favor at least picomolar amounts of the fusion molecule in the cell.
  • the entry and exit rates of the PTD and any cargo can be readily determined, or at least approximated, by standard kinetic analysis using detectably-labeled fusion molecules.
  • the ratio of the entry rate to the exit rate will be in the range of between about 5 to about 100 up to about 1000.
  • a PTD useful in the methods and compositions of the disclosure comprise a peptide featuring substantial alpha-helicity. It has been discovered that transduction is optimized when the PTD exhibits significant alpha-helicity.
  • the PTD comprises a sequence containing basic amino acid residues that are substantially aligned along at least one face of the peptide.
  • a PTD domain of the disclosure may be a naturally occurring peptide or a synthetic peptide.
  • the PTD comprises an amino acid sequences comprising a strong alpha helical structure with arginine (Arg) residues down the helical cylinder.
  • the PTD domain comprises a peptide represented by the following general formula: B 1 —X 1 —X 2 —X 3 —B 2 —X 4 —X 5 —B 3 (SEQ ID NO:1) wherein B 1 , B 2 , and B 3 are each independently a basic amino acid, the same or different; and X 1 , X 2 , X 3 , X 4 and X 5 are each independently an alpha-helix enhancing amino acid, the same or different.
  • the PTD domain is represented by the following general formula: B 1 —X 1 —X 2 —B 2 —B 3 —X 3 —X 4 —B 4 (SEQ ID NO:2) wherein B 1 , B 2 , B 3 , and B 4 are each independently a basic amino acid, the same or different; and X 1 , X 2 , X 3 , and X 4 are each independently an alpha-helix enhancing amino acid the same or different.
  • PTD domains comprise basic residues, e.g., lysine (Lys) or arginine (Arg), and further including at least one proline (Pro) residue sufficient to introduce “kinks” into the domain.
  • Examples of such domains include the transduction domains of prions.
  • such a peptide comprises KKRPKPG (SEQ ID NO:3).
  • the domain is a peptide represented by the following sequence: X—X—R—X—(P/X)—(B/X)—B—(P/X)—X—B—(B/X) (SEQ ID NO:4), wherein X is any alpha helical promoting residue such as alanine; P/X is either proline or X as previously defined; B is a basic amino acid residue, e.g., arginine (Arg) or lysine (Lys); R is arginine (Arg) and B/X is either B or X as defined above.
  • the PTD is cationic and consists of between 7 and 10 amino acids and has the formula K—X 1 —R—X 2 —X 1 (SEQ ID NO:5) wherein X 1 is R or K and X 2 is any amino acid.
  • An example of such a peptide comprises RKKRRQRRR (SEQ ID NO:6).
  • Additional transducing domains include a TAT fragment that comprises at least amino acids 49 to 56 of TAT up to about the full-length TAT sequence (see, e.g., SEQ ID NO:7).
  • a TAT fragment may include one or more amino acid changes sufficient to increase the alpha-helicity of the fragment.
  • the amino acid changes introduced will involve adding a recognized alpha-helix enhancing amino acid.
  • the amino acid changes will involve removing one or more amino acids from the TAT fragment that impede alpha helix formation or stability.
  • the TAT fragment will include at least one amino acid substitution with an alpha-helix enhancing amino acid.
  • a TAT fragment or other PTD will be made by standard peptide synthesis techniques although recombinant DNA approaches may be used in some cases.
  • Additional transduction proteins that can be used in the nucleic acid constructs of the disclosure include the TAT fragment in which the TAT 49-56 sequence has been modified so that at least two basic amino acids in the sequence are substantially aligned along at least one face of the TAT fragment.
  • Illustrative TAT fragments include at least one specified amino acid substitution in at least amino acids 49-56 of TAT which substitution aligns the basic amino acid residues of the 49-56 sequence along at least one face of the segment and typically the TAT 49-56 sequence.
  • Additional transduction proteins include the TAT fragment in which the TAT 49-56 sequence includes at least one substitution with an alpha-helix enhancing amino acid.
  • the substitution is selected so that at least two basic amino acid residues in the TAT fragment are substantially aligned along at least one face of that TAT fragment.
  • the substitution is chosen so that at least two basic amino acid residues in the TAT 49-56 sequence are substantially aligned along at least one face of that sequence.
  • chimeric PTD domains include parts of at least two different transducing proteins.
  • chimeric transducing proteins can be formed by fusing two different TAT fragments, e.g., one from HIV-1 and the other from HIV-2 or one from a prion protein and one from HIV.
  • PTDs can be linked or fused with any number of other molecules including an oligonucleotide or polynucleotide.
  • nucleic acid construct or PTD can be bound to other molecular entities including nucleic acid binding domains, targeting moieties and the like.
  • two or more PTDs e.g., 1-5, 2-4, typically 3
  • a nucleic acid binding domain can promote uptake of a fusion construct comprising a nucleic acid (including an oligonucleotide or polynucleotide comprising a protecting group) by reducing the anionic charge such that the cationic charge of the PTD domain is sufficient to transduce/traverse a cell's membrane.
  • RNA binding proteins include histone, RDE-4 protein, or protamine.
  • Additional dsRNA binding proteins include: PKR (AAA36409, AAA61926, Q03963), TRBP (P97473, AAA36765), PACT (AAC25672, AAA49947, NP609646), Staufen (AAD17531, AAF98119, AAD17529, P25159), NFAR1 (AF167569), NFAR2 (AF167570, AAF31446, AAC71052, AAA19960, AAA19961, AAG22859), SPNR (AAK20832, AAF59924, A57284), RHA (CAA71668, AAC05725, AAF57297), NREBP (AAK07692, AAF23120, AAF54409, T33856), kanadaptin (AAK29177, AAB88191, AAF55582, NP499172, NP198700, BAB19354), HYL1 (NP563850), hyponastic leaves (CAC05
  • two or more components of the constructs disclosed herein can be organized in nearly any fashion provided that the construct has the function for which it was intended (e.g., sufficiently cationic or having reduced anionic charge).
  • the constructs can include fusion polypeptides or chimeric proteins comprising one or more PTDs linked either directly or indirectly linked to a oligonucleotide or polynucleotide domain (e.g., a therapeutic or diagnostic DNA, RNA, siRNA and the like).
  • Each of the several domains may be directly linked or may be separated by a linker peptide.
  • the domains may be presented in any order.
  • the fusion polypeptides may include tags, e.g., to facilitate identification and/or purification of the fusion polypeptide, such as a 6 ⁇ HIS tag.
  • Peptide linkers that can be used in the fusion polypeptides and methods of the disclosure will typically comprise up to about 20 or 30 amino acids, commonly up to about 10 or 15 amino acids, and still more often from about 1 to 5 amino acids.
  • the linker sequence is generally flexible so as not to hold the fusion molecule in a single rigid conformation.
  • the linker sequence can be used, e.g., to space the PTD domain from the nucleic acid binding domain and/or nucleic acid domain.
  • the peptide linker sequence can be positioned between the protein transduction domain and the nucleic acid domain, e.g., to provide molecular flexibility.
  • the length of the linker moiety is chosen to optimize the biological activity of the polypeptide comprising a PTD domain fusion construct and can be determined empirically without undue experimentation.
  • the linker moiety should be long enough and flexible enough to allow a nucleic acid binding domain to freely interact with a nucleic acid or vice versa.
  • linker moieties are -Gly-Gly-, GGGGS (SEQ ID NO:8), (GGGGS)N (SEQ ID NO:9), GKSSGSGSESKS (SEQ ID NO:10), GSTSGSGKSSEGKG (SEQ ID NO:11), GSTSGSGKSSEGSGSTKG (SEQ ID NO:12), GSTSGSGKPGSGEGSTKG (SEQ ID NO:13), or EGKSSGSGSESKEF (SEQ ID NO:14).
  • Linking moieties are described, for example, in Huston et al., Proc. Nat'l Acad. Sci.
  • compositions, and fusion polypeptides of the disclosure provide enhanced uptake and release of nucleic acid molecules by cells both in vitro and in vivo.
  • therapeutic is used in a generic sense and includes treating agents, prophylactic agents, and replacement agents.
  • therapeutic molecules include, but are not limited to, cell cycle control agents; agents which inhibit cyclin proteins, such as antisense polynucleotides to the cyclin G1 and cyclin D1 genes; dsRNA that can be cleaved to provide siRNA molecules directed to specific growth factors such as, for example, epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), erythropoietin, G-CSF, GM-CSF, TGF- ⁇ , TGF- ⁇ , and fibroblast growth factor; cytokines, including, but not limited to, Interleukins 1 through 13 and tumor necrosis factors; anticoagulants, anti-platelet agents; TNF receptor domains etc.
  • EGF epidermal growth factor
  • VEGF vascular endothelial growth factor
  • cytokines including, but not limited to, Interleukins 1 through 13 and tumor necrosis factors
  • various diseases and disorders can be treated.
  • growth of tumor cells can be inhibited, suppressed, or destroyed upon delivery of an anti-tumor siRNA.
  • any anionically charged nucleic acid e.g., dsRNA, siRNA and the like
  • dsRNA dsRNA
  • siRNA siRNA
  • polypeptides used in the disclosure can comprise either the L-optical isomer or the D-optical isomer of amino acids or a combination of both.
  • Polypeptides that can be used in the disclosure include modified sequences such as glycoproteins, retro-inverso polypeptides, D-amino acid modified polypeptides, and the like.
  • a polypeptide includes naturally occurring proteins, as well as those which are recombinantly or synthetically synthesized.
  • “Fragments” are a portion of a polypeptide.
  • fragment refers to a portion of a polypeptide which exhibits at least one useful epitope or functional domain.
  • a functional fragment refers to fragments of a polypeptide that retain an activity of the polypeptide.
  • a functional fragment of a PTD includes a fragment which retains transduction activity.
  • Biologically functional fragments can vary in size from a polypeptide fragment as small as an epitope capable of binding an antibody molecule, to a large polypeptide capable of participating in the characteristic induction or programming of phenotypic changes within a cell.
  • An “epitope” is a region of a polypeptide capable of binding an immunoglobulin generated in response to contact with an antigen.
  • retro-inverso peptides are used. “Retro-inverso” means an amino-carboxy inversion as well as enantiomeric change in one or more amino acids (i.e., levantory (L) to dextrorotary (D)).
  • a polypeptide of the disclosure encompasses, for example, amino-carboxy inversions of the amino acid sequence, amino-carboxy inversions containing one or more D-amino acids, and non-inverted sequence containing one or more D-amino acids.
  • Retro-inverso peptidomimetics that are stable and retain bioactivity can be devised as described by Brugidou et al. (Biochem. Biophys. Res. Comm. 214(2): 685-693, 1995) and Chorev et al. (Trends Biotechnol. 13(10): 438-445, 1995).
  • the disclosure provides a method of producing a fusion polypeptide comprising a PTD domain, a nucleic acid binding domain (e.g., DRBD) and a nucleic acid molecule by growing a host cell comprising a polynucleotide encoding the fusion polypeptide under conditions that allow expression of the polynucleotide, and recovering the fusion polypeptide.
  • a polynucleotide encoding a fusion polypeptide of the disclosure can be operably linked to a promoter for expression in a prokaryotic or eukaryotic expression system.
  • such a polynucleotide can be incorporated in an expression vector to generate a fusion construct.
  • the disclosure also provides polynucleotides encoding a fusion protein construct of the disclosure.
  • Such polynucleotides comprise sequences encoding one or more PTD domains, and/or a nucleic acid binding domain (e.g., DRBD).
  • the polynucleotide may also encode linker domains that separate one or more of the PTDs and/or nucleic acid binding domains.
  • a fusion polypeptide comprising two or more PTD domains is produced and then linked to a charge reduced/protected oligonucleotide or polynucleotide.
  • Delivery of a polynucleotide of the disclosure can be achieved by introducing the polynucleotide into a cell using a variety of methods known to those of skill in the art.
  • a construct comprising such a polynucleotide can be delivered into a cell using a colloidal dispersion system.
  • a polynucleotide construct can be incorporated (i.e., cloned) into an appropriate vector.
  • the polynucleotide encoding a fusion polypeptide of the disclosure may be inserted into a recombinant expression vector.
  • the term “recombinant expression vector” refers to a plasmid, virus, or other vehicle known in the art that has been manipulated by insertion or incorporation of a polynucleotide encoding a fusion polypeptide of the disclosure.
  • the expression vector typically contains an origin of replication, a promoter, as well as specific genes that allow phenotypic selection of the transformed cells.
  • Vectors suitable for such use include, but are not limited to, the T7-based expression vector for expression in bacteria (Rosenberg et al., Gene, 56:125, 1987), the pMSXND expression vector for expression in mammalian cells (Lee and Nathans, J. Biol. Chem., 263:3521, 1988), baculovirus-derived vectors for expression in insect cells, cauliflower mosaic virus, CaMV, and tobacco mosaic virus, TMV, for expression in plants.
  • any of a number of suitable transcription and translation elements may be used in the expression vector (see, e.g., Bitter et al., Methods in Enzymology, 153:516-544, 1987). These elements are well known to one of skill in the art.
  • operably linked and “operably associated” are used interchangeably herein to broadly refer to a chemical or physical coupling of two otherwise distinct domains that each have independent biological function.
  • operably linked refers to the functional linkage between a regulatory sequence and the polynucleotide regulated by the regulatory sequence.
  • operably linked refers to the association of a nucleic acid domain and a transduction domain such that each domain retains its independent biological activity under appropriate conditions.
  • Operably linked further refers to the link between encoded domains of the fusion polypeptides such that each domain is linked in-frame to give rise to the desired polypeptide sequence.
  • yeast a number of vectors containing constitutive or inducible promoters may be used (see, e.g., Current Protocols in Molecular Biology, Vol. 2, Ed. Ausubel et al., Greene Publish. Assoc. & Wiley Interscience, Ch. 13, 1988; Grant et al., “Expression and Secretion Vectors for Yeast,” in Methods in Enzymology, Eds. Wu & Grossman, Acad. Press, N.Y., Vol. 153, pp. 516-544, 1987; Glover, DNA Cloning, Vol. II, IRL Press, Wash., D.C., Ch.
  • An expression vector can be used to transform a host cell.
  • transformation is meant a permanent genetic change induced in a cell following incorporation of a polynucleotide exogenous to the cell. Where the cell is a mammalian cell, a permanent genetic change is generally achieved by introduction of the polynucleotide into the genome of the cell.
  • transformed cell or “recombinant host cell” is meant a cell into which (or into an ancestor of which) has been introduced, by means of molecular biology techniques, a polynucleotide encoding a fusion polypeptide of the disclosure. Transformation of a host cell may be carried out by conventional techniques as are known to those skilled in the art.
  • competent cells which are capable of polynucleotide uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCl2 method by procedures known in the art.
  • CaCl2 MgCl2 or RbCl can be used. Transformation can also be performed after forming a protoplast of the host cell or by electroporation.
  • a fusion polypeptide of the disclosure can be produced by expression of polynucleotide encoding a fusion polypeptide in prokaryotes.
  • polynucleotide encoding a fusion polypeptide include, but are not limited to, microorganisms, such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA, or cosmid DNA expression vectors encoding a fusion polypeptide of the disclosure.
  • the constructs can be expressed in E. coli in large scale. Purification from bacteria is simplified when the sequences include tags for one-step purification by nickel-chelate chromatography.
  • a polynucleotide encoding a fusion polypeptide can also comprise a tag to simplify isolation of the fusion polypeptide.
  • a polyhistidine tag of, e.g., six histidine residues, can be incorporated at the amino terminal end of the fusion polypeptide.
  • the polyhistidine tag allows convenient isolation of the protein in a single step by nickel-chelate chromatography.
  • a fusion polypeptide of the disclosure can also be engineered to contain a cleavage site to aid in protein recovery the cleavage site may be part of a linker moiety as discussed above.
  • a DNA sequence encoding a desired peptide linker can be inserted between, and in the same reading frame as, a polynucleotide encoding a PTD, or fragment thereof followed by a nucleic acid binding domain, the PTD may also be linked to a desired nucleic acid (e.g., dsRNA, DNA, siRNA, and the like), using any suitable conventional technique.
  • a chemically synthesized oligonucleotide encoding the linker can be ligated between two coding polynucleotides.
  • a polynucleotide of the disclosure will encode a fusion polypeptide comprising from two to four separate domains (e.g., one or more PTD domain and one or more a nucleic acid domains) separated by linkers.
  • a fusion polypeptide comprising a plurality of PTDs is associated or linked with an oligonucleotide comprising a phosphodiester and/or phosphothioate protecting group or other anionic charge reducing group.
  • Eukaryotic cells When the host cell is a eukaryotic cell, such methods of transfection of DNA as calcium phosphate co-precipitates, conventional mechanical procedures, such as microinjection, electroporation, insertion of a plasmid encased in liposomes, or virus vectors may be used. Eukaryotic cells can also be cotransfected with a polynucleotide encoding the PTD-fusion polypeptide of the disclosure, and a second polynucleotide molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene.
  • a selectable phenotype such as the herpes simplex thymidine kinase gene.
  • Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the fusion polypeptide (see, e.g., Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982).
  • a eukaryotic viral vector such as simian virus 40 (SV40) or bovine papilloma virus
  • Eukaryotic systems and typically mammalian expression systems, allow for proper post-translational modifications of expressed mammalian proteins to occur.
  • Eukaryotic cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, phosphorylation, and advantageously secretion of the fusion product can be used as host cells for the expression of the PTD-fusion polypeptide of the disclosure.
  • host cell lines may include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, Jurkat, HEK-293, and WI38.
  • telomeres For long-term, high-yield production of recombinant proteins, stable expression is used.
  • host cells can be transformed with the cDNA encoding a fusion polypeptide of the disclosure controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, and the like), and a selectable marker.
  • expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, and the like
  • the selectable marker in the recombinant plasmid confers selectivity (e.g., by cytotoxin resistance) and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci that, in turn, can be cloned and expanded into cell lines.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • a number of selection systems may be used, including, but not limited to, the herpes simplex virus thymidine kinase (Wigler et al., Cell, 11:223, 1977), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci.
  • adenine phosphoribosyltransferase genes can be employed in tk-, hgprt- or aprt- cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate (Wigler et al., Proc. Natl. Acad. Sci. USA, 77:3567, 1980; O'Hare et al., Proc. Natl. Acad. Sci.
  • gpt which confers resistance to mycophenolic acid
  • neo which confers resistance to the aminoglycoside G-418
  • hygro which confers resistance to hygromycin genes
  • trpB which allows cells to utilize indole in place of tryptophan
  • hisD which allows cells to utilize histinol in place of histidine
  • ODC ornithine decarboxylase
  • DFMO 2-(difluoromethyl)-DL-ornithine
  • Techniques for the isolation and purification of either microbially or eukaryotically expressed PTD-fusion polypeptides of the disclosure may be by any conventional means, such as, for example, preparative chromatographic separations and immunological separations, such as those involving the use of monoclonal or polyclonal antibodies or antigen.
  • the fusion polypeptides of the disclosure are useful for the delivery of anionically charged nucleic acid molecules (e.g., dsRNA, siRNA, DNA, antisense, ribozymes and the like) for the treatment and/or diagnosis of a number of diseases and disorders.
  • the fusion polypeptides can be used in the treatment of cell proliferative disorders, wherein the protected oligo- or polynucleotide is reversibly modified such that it traverses a cell membrane alone or in associate with a PTD to target genes that induce cell proliferation.
  • the PTD domain increases the overall net cationic charge or reduces the overall net anionic charge of the nucleic acid construct facilitating facilitates uptake by the cell.
  • the constructs are useful for treatment of cells having cell proliferative disorders.
  • the constructs of the disclosure can be used to treat inflammatory diseases and disorders, infections, vascular disease and disorders and the like.
  • the construct of the disclosure may alternatively comprise, or in addition to, the PTD, a targeting domain.
  • the targeting domain can be a receptor, receptor ligand or antibody useful for directing the construct to a particular cell type that expresses the cognate binding domain.
  • a construct of the disclosure will be formulated with a pharmaceutically acceptable carrier, although the fusion polypeptide may be administered alone, as a pharmaceutical composition.
  • a pharmaceutical composition according to the disclosure can be prepared to include a fusion polypeptide of the disclosure, into a form suitable for administration to a subject using carriers, excipients, and additives or auxiliaries.
  • carriers or auxiliaries include magnesium carbonate, titanium dioxide, lactose, mannitol and other sugars, talc, milk protein, gelatin, starch, vitamins, cellulose and its derivatives, animal and vegetable oils, polyethylene glycols and solvents, such as sterile water, alcohols, glycerol, and polyhydric alcohols.
  • Intravenous vehicles include fluid and nutrient replenishers.
  • Preservatives include antimicrobial, anti-oxidants, chelating agents, and inert gases.
  • compositions include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like, as described, for instance, in Remington's Pharmaceutical Sciences, 15th ed., Easton: Mack Publishing Co., 1405-1412, 1461-1487 (1975), and The National Formulary XIV., 14th ed., Washington: American Pharmaceutical Association (1975), the contents of which are hereby incorporated by reference.
  • the pH and exact concentration of the various components of the pharmaceutical composition are adjusted according to routine skills in the art. See Goodman and Gilman's, The Pharmacological Basis for Therapeutics (7th ed.).
  • compositions according to the disclosure may be administered locally or systemically.
  • “therapeutically effective dose” is meant the quantity of a fusion polypeptide according to the disclosure necessary to prevent, to cure, or at least partially arrest the symptoms of a disease or disorder (e.g., to inhibit cellular proliferation). Amounts effective for this use will, of course, depend on the severity of the disease and the weight and general state of the subject. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of the pharmaceutical composition, and animal models may be used to determine effective dosages for treatment of particular disorders. Various considerations are described, e.g., in Langer, Science, 249: 1527, (1990); Gilman et al. (eds.) (1990), each of which is herein incorporated by reference.
  • administering a therapeutically effective amount is intended to include methods of giving or applying a pharmaceutical composition of the disclosure to a subject that allow the composition to perform its intended therapeutic function.
  • the therapeutically effective amounts will vary according to factors, such as the degree of infection in a subject, the age, sex, and weight of the individual. Dosage procedures can be adjusted to provide the optimum therapeutic response. For example, several divided doses can be administered daily or the dose can be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • the pharmaceutical composition can be administered in a convenient manner, such as by injection (e.g., subcutaneous, intravenous, and the like), oral administration, inhalation, transdermal application, or rectal administration.
  • the pharmaceutical composition can be coated with a material to protect the pharmaceutical composition from the action of enzymes, acids, and other natural conditions that may inactivate the pharmaceutical composition.
  • the pharmaceutical composition can also be administered parenterally or intraperitoneally.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof, and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the composition will typically be sterile and fluid to the extent that easy syringability exists.
  • the composition will be stable under the conditions of manufacture and storage and preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size, in the case of dispersion, and by the use of surfactants.
  • a coating such as lecithin
  • surfactants Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols, such as mannitol, sorbitol, or sodium chloride are used in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the pharmaceutical composition in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the pharmaceutical composition into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the pharmaceutical composition can be orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the pharmaceutical composition and other ingredients can also be enclosed in a hard or soft-shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • the pharmaceutical composition can be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 1% by weight of active compound.
  • the percentage of the compositions and preparations can, of course, be varied and can conveniently be between about 5% to about 80% of the weight of the unit.
  • the tablets, troches, pills, capsules, and the like can also contain the following: a binder, such as gum gragacanth, acacia, corn starch, or gelatin; excipients such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid, and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin, or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring.
  • a binder such as gum gragacanth, acacia, corn starch, or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid, and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin, or a flavoring agent such
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the pharmaceutical composition can be incorporated into sustained-release preparations and formulations.
  • a “pharmaceutically acceptable carrier” is intended to include solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like.
  • solvents dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the pharmaceutical composition, use thereof in the therapeutic compositions and methods of treatment is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of pharmaceutical composition is calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the disclosure are related to the characteristics of the pharmaceutical composition and the particular therapeutic effect to be achieve.
  • compositions containing supplementary active ingredients are compounded for convenient and effective administration in effective amounts with a suitable pharmaceutically acceptable carrier in an acceptable dosage unit.
  • dosages are determined by reference to the usual dose and manner of administration of the said ingredients.
  • Phenyl chloroformate is added to Guanidine hydrochloride under basic conditions. Selective ring opening of thirane is accomplished under lewis acidic conditions to provide the free thiol.
  • Thiolated material is linked to betamercaptoethanol via a disulfide linkage to provide a free alcohol. Reaction of the free alcohol with tetraisopropylphosphonamidic chloride provides the diamidite capable of reaction with the free 3′OH of any nucleoside amidite precursor.
  • Oligonucleotides were synthesized using techniques known in the art as described herein.
  • Reactions were carried out under an argon atmosphere. Glassware was cleaned overnight in a KOH/EtOH base bath, rinsed with MeOH and flame dried under vacuum before use in all anhydrous systems. Reactions were run with solvents that were either purchased sure-sealed over molecular sieves or were distilled using protocols listed in Purification of Laboratory Chemicals 4th ed. and stored over sieves.
  • Tetrahydrofuran was distilled from sodium metal and benzophenone, triethyl amine (Et 3 N), di-isopropyl amine (DIEA) and pyridine (py) were distilled from sodium metal.
  • Dichloromethane CH 2 Cl 2
  • MeOH methanol
  • toluene were distilled from calcium hydride. All other solvents and reagents were purchased from Fisher Chemical Co., Aldrich Chemical Co., EMD or Acros Organics and used without further purification. Reactions were cooled to ⁇ 78° C. via dry ice-acetone baths.
  • Flash column chromatography was performed using Merck grade 60 silica gel (230-400 mesh) and TLC analysis was carried out using Merck 60F-254 pre-coated silica sheets. Visualization of TLC plates was achieved using ultraviolet light, p-anisaldehyde in ethanol with sulfuric acid, polyphosphomolybdic acid and cerium sulfate in EtOH with H 2 SO 4 , ninhydrin in EtOH with H 2 SO 4 , potassium permanganate or iodine. Solvent removal was effected by Büchi rotary evaporator equipped with a dry ice isopropanol cold finger trap, and a H 2 O aspirator was used to concentrate in vacuo.
  • Oligonucleotides of 17-21 nt in length were synthesized on an MerMade 6 automated DNA/RNA synthesizer. Glen Research Q CPG support was used with ethylthioltetrazole as the coupling reagent during 3 coupling steps of 5 minutes each. Phosphine was oxidized to phosphate by the standard iodine method and capping was performed with phenoxyacetic anhydride. All amidites and materials used on the MerMade 6 synthesizer were either synthesized or purchased from EMD, ChemGenes or Glen Research.
  • nucleoside activation as an phosphotriester amidite for solid phase synthesis Nucleoside amidites were prepared in a manner suitable for the Caruthers method of solid phase oligonucleotide synthesis. Nucleoside and activator are added to a flame dried vial. Phosphoramidite is added to the stirring solution reaction was allowed to proceed for a minimum of 3 h. Solvent was removed from the reactions and the resulting foam or oil was applied directly to a triethylamine pretreated silica column in running buffer. Fractions containing only product were pooled and the solvent was removed. The resulting foam was dissolved in acetonitrile and syringe filtered (0.45 um).
  • Acetonitrile was removed and the resulting foam was dissolved in benzene and lyophilized to remove all traces of triethyl amine and residual water. Samples were sealed in glass containers under argon and stored at ⁇ 20° C. prior to use on the automated oligonucleoside synthesizer.
  • phenooxyacetal base protection was used during synthesis.
  • a hindered base for cleavage form the solid support, which did not use a photolabile linker was used.
  • One advantage of the disclosure is the ability to use either or both of DNA and RNA nucleotides.
  • nucleosides containing SPTE phosphine protecting groups 2′ F. nucleosides containing SPTE phosphine protecting groups:
  • Methylphosphonates of the general structure listed above also provide access to oligonucleotides with charge neutral backbones that additionally provide nuclease resistance.

Abstract

This disclosure relates to nucleic acid constructs modified to have a reduced net anionic charge. In some aspects the constructs comprise phosphodiester and/or phosphothioate protecting groups. The disclosure also provide methods of making and using such constructs.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application Ser. No. 60/830,571 filed Jul. 12, 2006, the disclosure of which is incorporated herein by reference.
  • TECHNICAL FIELD
  • This invention relates to compositions and methods for transducing cells.
  • BACKGROUND
  • Polyanionic oligomers do not readily diffuse across cell membranes. In order to overcome this problem for cultured cells, cationic lipids when combined with anionic oligonucleotides are generally used to assist uptake. Unfortunately, this complex is generally toxic to cells, which means that both the exposure time and concentration of cationic lipid must be carefully controlled to insure transfection of viable cells.
  • Nucleic acid delivery to cells both in vitro and in vivo has been performed using various recombinant viral vectors and electroporation. Such techniques have sought to treat various diseases and disorders by knocking-out gene expression or providing genetic constructions for gene therapy.
  • The discovery of RNA interference (RNAi) as a cellular mechanism that selectively degrades mRNAs allows for both the targeted manipulation of cellular phenotypes in cell culture and the potential for development of directed therapeutics (Behlke, Mol. Ther. 13, 644-670, 2006; Xie et al., Drug Discov. Today 11, 67-73, 2006). Although short interfering RNA's (siRNAs) have great potential for manipulation of cellular phenotypes, due to their size and negative (anionic) charged nature, siRNAs are macromolecules with no ability to enter cells. Indeed, siRNAs are 25× in excess of Lipinski's “Rule of 5s” for cellular delivery of membrane diffusible molecules that generally limits size to less than 500 Da. Consequently, in the absence of a delivery vehicle or transfection agent, naked siRNAs do not enter cells, even at millimolar concentrations (Barquinero et al., Gene Ther. 11 Suppl 1, S3-9, 2004). Significant attention has been focused on the use of cationic lipids that both condense the siRNA and punch holes in the cellular membrane to solve the siRNA delivery problem. Although widely used, transfection reagents fail to achieve efficient delivery into many cell types, especially primary cells and hematopoietic cell lineages (T and B cells, macrophage). Moreover, lipofection reagents often result in varying degrees of cytotoxicity ranging from mild in tumor cells to high in primary cells.
  • Recent cell-directed targeting approaches using antibody fusions to DNA-condensing protamine (Song et al., Nat. Biotechnol. 23, 709-717, 2005) and siRNA fusions to receptor targeted RNA aptamers (McNamara et al., Nat. Biotechnol. 24, 1005-1015, 2006) offer the potential to delivery siRNAs into select cells.
  • SUMMARY
  • The disclosure provides methods and compositions for delivering masked oligonucleotides or polynucleotides into living cells. Reduced anionically charged, neutral and cationically charged oligonucleotides or polynucleotides either alone or conjugated to a transduction molecule traverse cell membranes better than anionically charged nucleic acid molecules. Transiently protected oligonucleotides or polynucleotides comprising a protecting/charge neutralizing group (e.g., a cationic peptide conjugated neutral phosphotriester based oligonucleotides or polynucleotides and positively charged phosphotriester oligonucleotides or polynucleotides) can be synthesized by automated amidite methods. These compounds can enter the cytosol of living cells by endocytic or macropinocytic mechanisms. In one aspect, the phosphotriester protecting/neutralizing group when exposed to the intracellular environment is designed to be removed by enzymatic activity or by passive intracellular methods (e.g., high intracellular concentrations of glutathione) to give phosphodiester oligonucleotides or polynucleotides capable of eliciting an RNAi response. Accordingly, the disclosure provides oligonucleotide prodrugs useful as therapeutics, diagnostics and as tools for research.
  • The disclosure further provides a peptide RNA/DNA hybrid pro-drug where the phosphates that are normally found in a negatively charged state under physiological conditions along the backbone of an oligonucleotide or polynucleotide are transiently protected as a nonionic charge neutral phosphotriester. In one aspect, the charge neutralizing phosphotriester group is formed by the addition of any number of different phosphate protecting groups (e.g., S-pivaloyl thioethanol (SPTE) group). In an alternate embodiment, the phosphate backbone is protected with a biologically reversible R group that contains a positive charge, e.g., a guanidinium group via an enzymatically labile linker (RNB) to give net cationic character to the construct. The oligonucleotide pro-drug of the disclosure with a net positive charge, by way of peptide linkage or by synthetically adding positive charge to the phosphate backbone of the oligonucleotide, allows for cell membrane penetration.
  • The disclosure provides a nucleic acid construct comprising: a) an oligonucleotide or polynucleotide domain comprising a phosphodiester and/or phosphothioate protecting group that reduces the net anionic charge of the oligonucleotide or polynucleotide backbone; and b) a transduction domain comprising a membrane transport function operably linked to the oligonucleotide or polynucleotide domain. In one aspect, the phosphodiester and/or phosphothioate protecting group has the general formula:

  • R—X— or Q-X—
  • wherein X is O, S or NR1, and R1 is H, methyl, ethyl, S-pivaloyl thioethanol, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • wherein R2 is selected from the group consisting of:

  • R2,
  • wherein R2 is alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic,
  • Figure US20090093425A1-20090409-C00001
  • wherein R3 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A1 and A2 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00002
  • wherein R4 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A3 and A4 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00003
  • wherein R5 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A5 is a one to seven atom chain, or substituted one to seven atom chain,
  • wherein X1 and X2 are each independently O, S or NR7, and R7 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Figure US20090093425A1-20090409-C00004
  • wherein R6 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A6 and A6 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • wherein X3 and X4 are each independently O, S or NR8, and R8 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • wherein Q is selected from the group consisting of:

  • Q1,
  • wherein Q1 is a basic group with a pKa greater than or equal to 10,
  • Figure US20090093425A1-20090409-C00005
  • wherein Q2 is a basic group with a pKa greater than or equal to 10,
  • wherein A8 and A9 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00006
  • wherein Q3 is a basic group with a pKa greater than or equal to 10,
  • wherein A10 and A11 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00007
  • wherein Q4 is a basic group with a pKa greater than or equal to 10,
  • wherein A12 is a one to seven atom chain, or substituted one to seven atom chain,
  • wherein X5 and X6 are each independently O, S or NR9, and R9 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Figure US20090093425A1-20090409-C00008
  • wherein Q5 is a basic group with a pKa greater than or equal to 10,
  • wherein A13 and A14 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • wherein X7 and X8 are each independently O, S or NR10, and R10 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic. In another aspect the phosphodiester and/or phosphothioate protecting group is selected from the group consisting of MeO-, EtO-, iPrO,
  • Figure US20090093425A1-20090409-C00009
  • The disclosure also provides a nucleic acid construct comprising: an oligonucleotide or polynucleotide comprising a phosphodiester and/or phosphothioate protecting group that reduces the net anionic charge of the oligonucleotide or polynucleotide backbone or provides a net cationic charge of the oligonucleotide or polynucleotide backbone. In one aspect, the phosphodiester and/or phosphothioate protecting group has the general formula:

  • R—X— or Q-X—
  • wherein X is O, S or NR1, and R1 is H, methyl, ethyl, S-pivaloyl thioethanol, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • wherein R is selected from the group consisting of:

  • R2,
  • wherein R2 is alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic,
  • Figure US20090093425A1-20090409-C00010
  • wherein R3 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A1 and A2 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00011
  • wherein R4 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A3 and A4 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00012
  • wherein R5 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A5 is a one to seven atom chain, or substituted one to seven atom chain,
  • wherein X1 and X2 are each independently O, S or NR7, and R7 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Figure US20090093425A1-20090409-C00013
  • wherein R6 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A6 and A6 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • wherein X3 and X4 are each independently O, S or NR8, and R8 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • wherein Q is selected from the group consisting of:

  • Q1,
  • wherein Q1 is a basic group with a pKa greater than or equal to 10,
  • Figure US20090093425A1-20090409-C00014
  • wherein Q2 is a basic group with a pKa greater than or equal to 10,
  • wherein A8 and A9 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00015
  • wherein Q3 is a basic group with a pKa greater than or equal to 10,
  • wherein A10 and A11 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00016
  • wherein Q4 is a basic group with a pKa greater than or equal to 10,
  • wherein A12 is a one to seven atom chain, or substituted one to seven atom chain,
  • wherein X5 and X6 are each independently O, S or NR9, and R9 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Figure US20090093425A1-20090409-C00017
  • wherein Q5 is a basic group with a pKa greater than or equal to 10,
  • wherein A13 and A14 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • wherein X7 and X8 are each independently O, S or NR10, and R10 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic. In another aspect, the phosphodiester and/or phosphothioate protecting group is selected from the group consisting of MeO—, EtO—, iPrO,
  • Figure US20090093425A1-20090409-C00018
  • The disclosure also provides pharmaceutical composition comprising the nucleic acid constructs described herein.
  • The disclosure describes a method comprising linking one or more protein transduction domain to a nucleic acid construct. In one aspect, the one or more protein transduction domains comprise 2-5 protein transduction domains.
  • The disclosure also provides a method of generating a nucleic acid construct comprising: substantially purifying a protein transduction domain; synthesizing an oligonucleotide; charge neutralizing the anionic charge on the oligonucleotide with a phosphotriester group; and linking the oligonucleotide to one or more protein transduction domains.
  • Also provided are methods of transfecting a cell, comprising contacting the cell with a nucleic acid construct of the disclosure. The contacting can be in vivo or in vitro. The nucleic acid construct can comprise an antisense molecule.
  • The disclosure also provides a method of treating a disease or disorder comprising administering a nucleic acid construct of the disclosure to a subject, wherein the oligonucleotide or polynucleotide comprises a therapeutic or diagnostic molecule.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows TAT and anionically charged oligonucleotide interactions. TAT linked oligonucleotides lacking a phosphodiester and/or phosphothioate protecting group form complexes wherein the cationic charges are neutralized.
  • FIG. 2 depicts charge-neutralized oligonucleotides stimulating macropinocytosis.
  • FIG. 3 shows a general scheme of phosphotriester protection and charge neutralization during oligonucleotide uptake.
  • FIG. 4 show an embodiment of a PTD conjugated protected oligonucleotide and reversible deprotection.
  • FIG. 5 shows an embodiment a phosphodiester and/or phosphothioate protecting group formula.
  • FIG. 6 shows a nucleoside phosphoramidite synthesis route of the disclosure.
  • FIG. 7 depicts a reduction reaction for reversible charge neutralization.
  • FIG. 8 depicts an RNB phosphotriester protection and deprotection of anionic charged nucleic acids.
  • FIG. 9 shows chromatographic DTT reduction of a DTG protected uracil.
  • FIG. 10 shows redox sensitivity of DTG-Uridine phosphoramidite.
  • FIG. 11 shows dimer coupling reaction of DTG-Uridine phosphoramidite to di-TBS uridine.
  • FIG. 12 shows synthesis of poly-uridine oligos (SEQ ID NO:22) containing 2′ fluoride insertions with wildtype phosphodiester, methylphosphotriesters or SPTE-phosphotriesters. This figure shows the ability to synthesize RNA oligonucleotides containing phosphotriesters group with a 2′-Fluoro (F). Gel analysis performed under urea denaturing conditions followed by ethidium bromide staining (right panel).
  • FIG. 13A-D shows synthesis of double stranded siRNAs (SEQ ID NOs: 23-24) containing phosphotriester modifications. A: Structure of wild type phosphodiester and phosphotriester groups, methyl and SPTE are reversible, whereas the isopropyl group is not reversible and acts as a control. B: Sequence of eGFP siRNAs showing wild type Sense (S) strand (top line in panel) and Anti-Sense (AS) (also termed guide strand) (bottom line in panel) synthesized with six DNA thymidine insertions containing either wildtype, methyl, isopropyl or SPTE phosphotriesters as indicated. C: Single stranded RNA analysis of full length Anti-Sense (AS) oligonucleotides containing indicated phosphotriester modifications. Gel analysis performed under urea denaturing conditions followed by ethidium bromide staining. D: Double stranded RNA analysis of synthetic duplexes containing phosphotriester modifications. Sense (S) strand (top line) is wild type RNA. Anti-Sense (AS) strand (bottom line) contains 6× wild type DNA Thymidines (AS-6T), 6× methyl-phosphotriester Thymidines (AS-Met6T), 6× isopropyl-phosphotriester Thymidines (AS-Iso6T), 6× SPTE-phosphotriester Thymidines (AS-SPTE6T), as indicated. Gel analysis performed under non-denaturing conditions followed by ethidium bromide staining. This data shows the ability to make stable, full length siRNAs containing phosphotriester modifications.
  • FIG. 14A-B shows analysis of GFP reporter RNA Interference (RNAi) response. A: Flow cytometry (FACS) of reporter H1299 cells stably expressing eGFP following treatment with indicated siRNA duplexes. Cells were transfected with 50 nM indicated siRNAs plus Lipofectamine 2000 for 4 hr then analyzed for GFP expression by FACS at 24 hr. Untreated control cells show high level of GFP expression. Transfection of irreversible S/AS-Iso6T RNA showed no change in GFP expression, indicating a blocking or failure to induce an RNAi response by the continued presence of the 6× isopropyl phosphotriester groups. This observation demonstrates the requirement for phosphotriesters to reverse (decompose) to phosphodiesters to induce a RNAi response. Importantly, transfection of either S/AS-Met6T or S/AS-SPTE6T siRNAs induced significant RNAi responses, respectively, that were close to wild type control A/AS-6T (grey) RNAi response. These observations demonstrate the use of phosphodiesters on the Anti-Sense (guide) strand and the ability of the reversible Met6T and SPTE6T phosphotriesters groups to decompose inside the cell prior to being loaded into the RNAi machinery. B: Dose curve of indicated siRNAs from A showing RNAi efficiency at various concentrations. H1299 expressing GFP reporter cells were treated for 4 hours with siRNA plus Lipofectamine 2000. RNAi efficiency was analyzed at 24 hours post-transfection by FACS.
  • FIG. 15A-B shows PTD mediated delivery of charge neutralized, dsDNA. A: dsDNA containing 15× charge neutral Methyl-phosphotriesters (15N) on each strand and containing a a thiol crosslinking motif on the 5′ end of the Sense (S) strand and a Cy3 dye label on the 5′ end of the Anti-Sense (AS) strand were were chemically cross-linked to either 2×TAT PTDs (left panel) or 3×TAT-PTDs (right panel) as indicated, purified as dsDNA molecules, added to cells for 4 hr, trypsinized, washed before analysis of siRNA uptake via Cy3 label by flow cytometry (FACS). dsDNA is only taken up when conjugated to either 2× or 3×PTD peptides via a disulfide bond (2×/3×PTD-S—S-15N/15N-Cy3), whereas control of peptide added to dsDNA, but NOT conjugated (2×/3×PTD+15N/15N-Cy3), is not taken up by cells. B: Dose curve graph of PTD-dsDNA-Cy3 oligo conjugates from above after 4 hour treatment of H1299 cells by FACS as indicated.
  • DETAILED DESCRIPTION
  • As used herein and in the appended claims, the singular forms “a,” “and,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a PTD” includes a plurality of such PTDs and reference to “the cell” includes reference to one or more cells known to those skilled in the art, and so forth.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this disclosure belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice of the disclosed methods and compositions, the exemplary methods, devices and materials are described herein.
  • The publications discussed above and throughout the text are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior disclosure.
  • The ability to deliver certain bioactive agents to the interior of cells is problematical due to the bioavailability restriction imposed by the cell membrane. The plasma membrane of the cell forms a barrier that restricts the intracellular uptake of molecules to those which are sufficiently non-polar and smaller than approximately 500 daltons in size. Previous efforts to enhance the cellular internalization of proteins have focused on fusing proteins with receptor ligands (Ng et al., Proc. Natl. Acad. Sci. USA, 99:10706-11, 2002) or by packaging them into caged liposomal carriers (Abu-Amer et al., J. Biol. Chem. 276:30499-503, 2001). However, these techniques often result in poor cellular uptake and intracellular sequestration into the endocytic pathway.
  • Due to their anionic charge and large size of ˜14,000 Daltons, delivery of siRNA is a formidable challenge in mammals, including humans. The ability to deliver siRNA in vivo via protein transduction domains opens up the potential treatment of many diseases. The disclosure provides methods and compositions to facilitate and improve cellular uptake of nucleic acid molecules by “protecting” the charge associated with an oligonucleotide or polynucleotide.
  • Other highly charged nucleic acid molecules with therapeutic potential face the same delivery barrier. For example, RNA aptamers have great potential to bind to, sequester and inhibit proteins, but at >10,000 Daltons and highly charged, they have no or limited ability to enter cells on their own. The methods and compositions of the disclosure allow for intracellular delivery of RNA aptamers, siRNA and DNA vectors.
  • Thus, the potential of sequence specific oligonucleotides or polynucleotides to selectively treat human diseases can more effectively be delivering useful oligonucleotides and polynucleotides, including siRNAs, RNA aptamers, and DNA vectors to subjects and to cells. The disclosure overcomes size and charge limitations that make such RNAi constructs difficult to deliver or undeliverable. By reversibly neutralizing the anionic charge on a nucleic acids (e.g., dsRNA), a construct comprising a phosphodiester and/or phosphothioate protecting group according to the disclosure can deliver nucleic acids into a cell in vitro and in vivo.
  • The disclosure provides nucleic acid constructs comprising phosphodiester and/or phosphothioate protecting groups. The construct can further include compositions useful in cellular transduction and cellular modulation. Such compositions can include transduction moiety domains comprising a membrane transport function and may further comprise a nucleic acid binding domain sufficient to reversibly neutralize anionic charges on nucleic acids.
  • For example, charge neutralization of anionic nucleic acid (e.g., an RNA molecule) using a phosphodiester and/or phosphothioate protecting group(s) promotes uptake. In embodiments where the charge neutralized anionic nucleic acid is linked to a PTD the charge neutralization of the anionic charged nucleic acid frees the cationic PTD to traverse the membrane as well as prevents aggregation of the conjugate. The exposed free cationic charge of the PTD can then effectively interact with a cell surface, induce macropinocytosis and escape from the macropinosome into the cytoplasm. Once inside a cell, the phosphodiester and/or phosphothioate protecting group(s) can be removed by intracellular processes, such as reduction of a disulfide linkage or ester hydrolysis, allowing for removal from the construct in the cytoplasm. A nucleic acid construct that includes, for example, dsRNA can then be hydrolyzed by Dicer, an RNAse III-like ribonuclease, thereby releasing siRNA that silences a target gene.
  • The data provided herein demonstrate that the addition of one or more removable (e.g., reversibly attached) phosphodiester and/or phosphothioate protecting group(s) to a nucleic acid can effectively facilitate cell transduction. Any nucleic acid, regardless of sequence composition, can be modified by phosphodiester and/or phosphothioate protecting group(s).
  • The disclosure provides oligonucleotides or polynucleotides having, in some embodiments, one or more bioreversible protecting groups that contribute to chemical and biophysical properties that enhance cellular membrane penetration and resistance to exo- and endonuclease degradation. The disclosure further provided amidite reagents for the synthesis of the bioreversible protected oligonucleotides or polynucleotides. Moreover, these protecting groups are stable during the synthetic processes.
  • The oligonucleotides or polynucleotides of the disclosure having one or more bioreversible protecting groups are sometimes referred to as pro-oligonucleotides or pro-polynucleotides. In embodiments of this disclosure, the pro-oligonucleotides are capable of improved cellular lipid bilayers penetrating potential as well as resistance to exo- and endonuclease degradation in vivo. In cells, the bioreversible protecting groups are removed in the cell cytosol by reducing conditions, enzymatic activity (e.g., endogenous carboxyesterases) and the like to yield biologically active oligonucleotide compounds that are capable of hybridizing to and/or having an affinity for specific endogenous nucleic acids.
  • The phosphodiester and/or phosphothioate protecting groups can be used with antisense oligonucleotides of synthetic DNA or RNA or mixed molecules of complementary sequences to a target sequence belonging to a gene or to an RNA messenger whose expression they are specifically designed to block or down-regulate. The antisense oligonucleotides may be directed against a target messenger RNA sequence or, alternatively against a target DNA sequence, and hybridize to the nucleic acid to which they are complementary. Accordingly, these molecules effectively block or down-regulate gene expression.
  • Protected oligonucleotides or polynucleotides may also be directed against certain bicatenary DNA regions (homopurine/homopyrimidine sequences or sequences rich in purines/pyrimidines) and thus form triple helices. The formation of a triple helix, at a particular sequence, can block the interaction of protein factors which regulate or otherwise control gene expression and/or may facilitate irreversible damage to be introduced to a specific nucleic acid site if the resulting oligonucleotide is made to possess a reactive functional group.
  • Provided herein are nucleic acid constructs, and methods of producing such constructs, that can be used for facilitating the delivery of oligonucleotides or polynucleotides in to cells. In one embodiment, a nucleic acid construct includes one or more phosphodiester and/or phosphothioate protecting group(s) to neutralize the phosphodiester anionic charge associated with a nucleic acid, such as RNA and/or DNA. Once inside the cell, the protecting group can be removed from the construct by intracellular processes that include disulfide linkage reduction, ester hydrolysis or other enzyme-mediated processes. In other embodiments, the nucleic acid construct comprising one or more phosphodiester and/or phosphothioate protecting group further comprises one or more transduction domains such as a protein transduction domain (PTD). For example, a PTD can be conjugated directly to an oligonucleotide (e.g., an RNA or DNA) comprising the nucleic acid construct, such as at the 5′ and/or 3′ end via a free thiol group. For example, a PTD can be linked to the construct by a biologically sensitive and reversible manner, such as a disulfide linkage. This approach can be applied to any oligonucleotide or polynucleotide length and will allow for delivery of RNA (e.g., siRNA, RNA apatmer) or DNA into cells.
  • In another embodiment, a nucleic acid construct can include a basic group, such as guanidium group (similar to the head group arginine, an active component of the PTD), linked to the reversible protecting group and thereby limit the need for the PTD.
  • Accordingly, provided herein are nucleic acid constructs synthesized to include phosphodiester and/or phosphothioate protecting group(s) for the delivery of nucleic acid sequences across a cell membrane. The construct can also include, for example, one or more transduction domains and/or a protecting group that contains a basic group. Once inside the cell the oligonucleotide/polynucleotide of the nucleic acid construct reverts to an unprotected/wild type oligonucleotide/polynucleotide based on the reducing environment, by hydrolysis or other enzymatic activity.
  • An isolated nucleic acid construct refers to an oligonucleotide or polynucleotide associated with a molecule or compound comprising a phosphodiester and/or phosphothioate protecting group. For example, a nucleic acid construct includes, but is not limited to, an oligonucleotide or polynucleotide associated with an anionic charge reducing group(s) or molecule(s), by hydrogen bonding, charge association, covalent bonding and the like, to promote uptake by a cell. Examples of anionic charge reducing molecules that can be associated with an oligonucleotide or polynucleotide in the nucleic acid constructs of the disclosure include fusion polypeptides or peptides, chemical moieties that reduce the net anionic charge of an oligonucleotide or polynucleotide and combinations thereof.
  • Applications for the nucleic acid constructs provided herein include selective treatment of cancer, viral infection, genetic diseases, nucleic acid delivery for research and the like.
  • The term polynucleotide(s) and oligonucleotide(s) generally refers to any polyribonucleotide or polydeoxyribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. Thus, for instance, an oligonucleotide as used herein refers to, among others, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions. Thus, a oligonucleotide can comprise an siRNA, an antisense molecule, a ribozyme and the like.
  • In addition, a polynucleotide or oligonucleotides also includes triple-stranded regions comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules.
  • In some aspects a polynucleotide or oligonucleotide includes DNAs or RNAs as described above that contain one or more modified bases. Thus, DNAs or RNAs with backbones comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, are polynucleotides or oligonucleotides as the term is used herein.
  • One aspect of the disclosure comprises a nucleic acid construct comprising an oligonucleotide with reduced anionic charged and including a phosphodiester and/or phosphothioate protecting group or charge neutralization group. However, the construct can further include transduction domains and/or nucleic acid binding domains. These approaches could include engineering a disulfide bond (or ester linkage, etc) between a nucleic acid construct (e.g., an siRNA comprising a phosphodiester protection group) and a protein transduction domain (PTD) or a PCT and a double stranded RNA binding domain (DRBD) (e.g., TAT-DRBD) fusion protein to further increase the binding avidity, uptake, anionic charge reduction, and/or increase cationic charge. Once taken up by a cell (in vitro or in vivo), the complex can be reduced (e.g., by disulfide reduction) and released inside the cell.
  • As used herein an anionic charge neutralizing molecule or group refers to a molecule or chemical group that can reduce the overall net anionic charge of an oligonucleotide or polynucleotide to which it is associated. Phosphodiester and/or phosphothioate protecting groups as described herein are anionic charge neutralizing groups. The phosphodiester and/or phosphothioate protecting groups can be reversible or irreversible. One or more anionic charge neutralizing molecules or groups can be associated with an oligonucleotide or polynucleotide wherein each independently contributes to a reduction or the anionic charge and or increase in cationic charge of the construct. For example, one or more phosphodiester and/or phosphothioate protecting groups can be associated with an oligonucleotide and the “protected oligonucleotide” associated with one or more cationic transduction domains (e.g., PTDs), such that the overall net anionic charge of the construct is reduced or the overall net charge of the construct is neutral or the overall net charge of the construct is cationic relative to the oligonucleotide without the phosphodiester and/or phosphothioate protecting group and/or PTD.
  • The disclosure provides phosphodiester and/or phosphothioate protecting groups and oligonucleotides or polynucleotides comprising such phosphodiester and/or phosphothioate protecting groups. In one embodiment, the phosphodiester and/or phosphothioate protecting group has the general formula:

  • R—X— or Q-X—
  • wherein X is O, S or NR1, and R1 is H, methyl, ethyl, S-pivaloyl thioethanol, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • wherein R is selected from the group consisting of:

  • R2,
  • wherein R2 is alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic,
  • Figure US20090093425A1-20090409-C00019
  • wherein R3 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A1 and A2 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00020
  • wherein R4 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A3 and A4 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00021
  • wherein R5 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A5 is a one to seven atom chain, or substituted one to seven atom chain,
  • wherein X1 and X2 are each independently O, S or NR7, and R7 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Figure US20090093425A1-20090409-C00022
  • wherein R6 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
  • wherein A6 and A6 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • wherein X3 and X4 are each independently O, S or NR8, and R8 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
  • wherein Q is selected from the group consisting of:

  • Q1,
  • wherein Q1 is a basic group with a pKa greater than or equal to 10,
  • Figure US20090093425A1-20090409-C00023
  • wherein Q2 is a basic group with a pKa greater than or equal to 10,
  • wherein A8 and A9 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00024
  • wherein Q3 is a basic group with a pKa greater than or equal to 10,
  • wherein A10 and A11 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • Figure US20090093425A1-20090409-C00025
  • wherein Q4 is a basic group with a pKa greater than or equal to 10,
  • wherein A12 is a one to seven atom chain, or substituted one to seven atom chain,
  • wherein X5 and X6 are each independently O, S or NR9, and R9 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
  • Figure US20090093425A1-20090409-C00026
  • wherein Q5 is a basic group with a pKa greater than or equal to 10,
  • wherein A13 and A14 are each independently one to seven atom chains, or substituted one to seven atom chains,
  • wherein X7 and X8 are each independently O, S or NR10, and R10 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic.
  • In another embodiment, the phosphodiester and/or phosphothioate protecting group is selected from the group consisting of MeO—, EtO—, iPrO,
  • Figure US20090093425A1-20090409-C00027
  • In yet another embodiment, the phosphodiester and/or phosphothioate protecting group comprises a structure selected from the group consisting of:
  • Figure US20090093425A1-20090409-C00028
  • In another embodiment, the RNB protecting group comprises a structure selected from the group consisting of:
  • Figure US20090093425A1-20090409-C00029
  • Alkyl groups include straight-chain, branched and cyclic alkyl groups. Alkyl groups include those having from 1 to 20 carbon atoms. Alkyl groups include small alkyl groups having 1 to 3 carbon atoms. Alkyl groups include medium length alkyl groups having from 4-10 carbon atoms. Alkyl groups include long alkyl groups having more than 10 carbon atoms, particularly those having 10-20 carbon atoms. Cyclic alkyl groups include those having one or more rings. Cyclic alkyl groups include those having a 3-, 4-, 5-, 6-, 7-, 8-, 9- or 10-member carbon ring and particularly those having a 3-, 4-, 5-, 6-, or 7-member ring. The carbon rings in cyclic alkyl groups can also carry alkyl groups. Cyclic alkyl groups can include bicyclic and tricyclic alkyl groups. Alkyl groups optionally include substituted alkyl groups. Substituted alkyl groups include among others those which are substituted with aryl groups, which in turn can be optionally substituted. Specific alkyl groups include methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, n-butyl, s-butyl, t-butyl, cyclobutyl, n-pentyl, branched-pentyl, cyclopentyl, n-hexyl, branched hexyl, and cyclohexyl groups, all of which are optionally substituted.
  • Alkenyl groups include straight-chain, branched and cyclic alkenyl groups. Alkenyl groups include those having 1, 2 or more double bonds and those in which two or more of the double bonds are conjugated double bonds. Alkenyl groups include those having from 2 to 20 carbon atoms. Alkenyl groups include small alkyl groups having 2 to 3 carbon atoms. Alkenyl groups include medium length alkenyl groups having from 4-10 carbon atoms. Alkenyl groups include long alkenyl groups having more than 10 carbon atoms, particularly those having 10-20 carbon atoms. Cyclic alkenyl groups include those having one or more rings. Cyclic alkenyl groups include those in which a double bond is in the ring or in an alkenyl group attached to a ring. Cyclic alkenyl groups include those having a 3-, 4-, 5-, 6-, 7-, 8-, 9- or 10-member carbon ring and particularly those having a 3-, 4-, 5-, 6- or 7-member ring. The carbon rings in cyclic alkenyl groups can also carry alkyl groups. Cyclic alkenyl groups can include bicyclic and tricyclic alkyl groups. Alkenyl groups are optionally substituted. Substituted alkenyl groups include among others those which are substituted with alkyl or aryl groups, which groups in turn can be optionally substituted. Specific alkenyl groups include ethenyl, prop-1-enyl, prop-2-enyl, cycloprop-1-enyl, but-1-enyl, but-2-enyl, cyclobut-1-enyl, cyclobut-2-enyl, pent-1-enyl, pent-2-enyl, branched pentenyl, cyclopent-1-enyl, hex-1-enyl, branched hexenyl, cyclohexenyl, all of which are optionally substituted.
  • Aryl groups include groups having one or more 5- or 6-member aromatic or heteroaromatic rings. Aryl groups can contain one or more fused aromatic rings. Heteroaromatic rings can include one or more N, O, or S atoms in the ring. Heteroaromatic rings can include those with one, two or three N, those with one or two 0, and those with one or two S. Aryl groups are optionally substituted. Substituted aryl groups include among others those which are substituted with alkyl or alkenyl groups, which groups in turn can be optionally substituted. Specific aryl groups include phenyl groups, biphenyl groups, pyridinyl groups, and naphthyl groups, all of which are optionally substituted.
  • Arylalkyl groups are alkyl groups substituted with one or more aryl groups wherein the alkyl groups optionally carry additional substituents and the aryl groups are optionally substituted. Specific alkylaryl groups are phenyl-substituted alkyl groups, e.g., phenylmethyl groups.
  • Alkylaryl groups are aryl groups substituted with one or more alkyl groups wherein the alkyl groups optionally carry additional substituents and the aryl groups are optionally substituted. Specific alkylaryl groups are alkyl-substituted phenyl groups such as methylphenyl.
  • The rings that may be formed from two or more of R1-R5 together can be optionally substituted cycloalkyl groups, optionally substituted cycloalkenyl groups or aromatic groups. The rings may contain 3, 4, 5, 6, 7 or more carbons. The rings may be heteroaromatic in which one, two or three carbons in the aromatic ring are replaced with N, O or S. The rings may be heteroalkyl or heteroalkenyl, in which one or more CH2 groups in the ring are replaced with O, N, NH, or S.
  • Optional substitution of any alkyl, alkenyl and aryl groups includes substitution with one or more of the following substituents: halogens, —CN, —COOR, —OR, —COR, —OCOOR, —CON(R) 2, —OCON(R) 2, —N(R) 2, —NO2, —SR, —SO2R, —SO2N(R) 2 or —SOR groups. Optional substitution of alkyl groups includes substitution with one or more alkenyl groups, aryl groups or both, wherein the alkenyl groups or aryl groups are optionally substituted. Optional substitution of alkenyl groups includes substitution with one or more alkyl groups, aryl groups, or both, wherein the alkyl groups or aryl groups are optionally substituted. Optional substitution of aryl groups includes substitution of the aryl ring with one or more alkyl groups, alkenyl groups, or both, wherein the alkyl groups or alkenyl groups are optionally substituted.
  • Optional substituents for alkyl, alkenyl and aryl groups include among others:
  • —COOR where R is a hydrogen or an alkyl group or an aryl group and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl groups all of which are optionally substituted; —
  • —COR where R is a hydrogen, or an alkyl group or an aryl groups and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted;
  • —CON(R)2 where each R, independently of each other R is a hydrogen or an alkyl group or an aryl group and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted; R and R can form a ring which may contain one or more double bonds;
  • —OCON(R)2 where each R, independently of each other R, is a hydrogen or an alkyl group or an aryl group and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted; R and R can form a ring which may contain one or more double bonds;
  • —N(R)2 where each R, independently of each other R, is a hydrogen, or an alkyl group, acyl group or an aryl group and more specifically where R is methyl, ethyl, propyl, butyl, or phenyl or acetyl groups all of which are optionally substituted; or R and R can form a ring which may contain one or more double bonds.
  • —SR, —SO2R, or —SOR where R is an alkyl group or an aryl groups and more specifically where R is methyl, ethyl, propyl, butyl, phenyl groups all of which are optionally substituted; for —-SR, R can be hydrogen;
  • —OCOOR where R is an alkyl group or an aryl groups;
  • —SO2N(R)2 where R is a hydrogen, an alkyl group, or an aryl group and R and R can form a ring;
  • —OR where R═H, alkyl, aryl, or acyl; for example, R can be an acyl yielding—OCOR* where R* is a hydrogen or an alkyl group or an aryl group and more specifically where R* is methyl, ethyl, propyl, butyl, or phenyl groups all of which groups are optionally substituted.
  • Specific substituted alkyl groups include haloalkyl groups, particularly trihalomethyl groups and specifically trifluoromethyl groups. Specific substituted aryl groups include mono-, di-, tri, tetra- and pentahalo-substituted phenyl groups; mono-, di-, tri-, tetra-, penta-, hexa-, and hepta-halo-substituted naphthalene groups; 3- or 4-halo-substituted phenyl groups, 3- or 4-alkyl-substituted phenyl groups, 3- or 4-alkoxy-substituted phenyl groups, 3- or 4-RCO-substituted phenyl, 5- or 6-halo-substituted naphthalene groups. More specifically, substituted aryl groups include acetylphenyl groups, particularly 4-acetylphenyl groups; fluorophenyl groups, particularly 3-fluorophenyl and 4-fluorophenyl groups; chlorophenyl groups, particularly 3-chlorophenyl and 4-chlorophenyl groups; methylphenyl groups, particularly 4-methylphenyl groups, and methoxyphenyl groups, particularly 4-methoxyphenyl groups.
  • The use of basic containing groups (e.g., guanidium groups) linked to reversible phosphodiester and/or phosphothioate protecting groups has not been used for nucleic acid delivery. Nor have others contemplated the use of dsRNA with protecting groups and/or the use of PTDs conjugated to oligonucleotides or polynucleotides comprising a charge neutralizing group for delivery of a nucleic acid construct across a cell membrane.
  • Where an oligonucleotide or polynucleotide is linked to a PTD, charge neutralization of the anionically charged oligonucleotide or polynucleotide frees the cationically charged PTD to productively interact with the cell surface and also prevents aggregation of the conjugate. For example, the exposed cationic charged PTD interacts with the cell surface and induces macropinocytosis. The oligonucleotide is released into the cytoplasm. Once inside the cell, the protecting group can be cleaved off by cellular processes, such as a reducing enzyme, oxidizing enzyme, reducing agent, oxidizing agent or esterase, unprotecting the oligonucleotide or polynucleotide allowing the nucleic acid to revert to its natural configuration.
  • As used herein, a nucleic acid domain, used interchangeably with oligonucleotide or polynucleotide domain, can be any oligonucleotide or polynucleotide (e.g., a ribozyme, antisense molecule, polynucleotide, oligonucleotide and the like). Oligonucleotides or polynucleotides generally contain phosphodiester bonds, although in some cases, nucleic acid analogs are included that may have alternate backbones, comprising, e.g., phosphoramidate, phosphorothioate, phosphorodithioate, or O-methylphophoroamidite linkages (see Eckstein, Oligonucleotides and Analogues: A Practical Approach, Oxford University Press); and peptide nucleic acid backbones and linkages. Other analog nucleic acids include those with positive backbones; non-ionic backbones, and non-ribose backbones, including those described in U.S. Pat. Nos. 5,235,033 and 5,034,506, and Chapters 6 and 7, ASC Symposium Series 580, Carbohydrate Modifications in Antisense Research, Sanghui & Cook, eds. Nucleic acids containing one or more carbocyclic sugars are also included within one definition of nucleic acids. Modifications of the ribose-phosphate backbone may be done for a variety of reasons, e.g. to increase the stability and half-life of such molecules in physiological environments. Mixtures of naturally occurring nucleic acids and analogs are encompassed by the term oligonucleotide and polynucleotide; alternatively, mixtures of different nucleic acid analogs, and mixtures of naturally occurring nucleic acids and analogs can be made. Furthermore, hybrids of RNN, RNB, DNA, and RNA can be used. dsDNA, ssDNA, dsRNA, siRNA are encompassed by the term oligonucleotide and polynucleotide.
  • A polynucleotide refers to a polymeric compound made up of any number of covalently bonded nucleotide monomers, including nucleic acid molecules such as DNA and RNA molecules, including single- double- and triple-stranded such molecules, and is expressly intended to embrace that group of polynucleotides commonly referred to as “oligonucleotides”, which are typically distinguished as having a relatively small number (no more than about 30, e.g., about 5-10, 10-20 or 20-30) of nucleotide constituents.
  • As used herein, the term “siRNA” is an abbreviation for “short interfering RNA”, also sometimes known as “small interfering RNA” or “silencing RNA”, and refers to a class of about 19-25 nucleotide-long double-stranded ribonucleic acid molecules that in eukaryotes are involved in the RNA interference (RNAi) pathway that results in post-transcriptional, sequence-specific gene silencing.
  • The term “dsRNA” is an abbreviation for “double-stranded RNA” and as used herein refers to a ribonucleic acid molecule having two complementary RNA strands and which stands distinct from siRNA in being at least about 26 nucleotides in length, and more typically is at least about 50 to about 100 nucleotides in length.
  • As described above, the nucleic acid may be DNA, both genomic and cDNA, RNA or a hybrid, where the nucleic acid may contain combinations of deoxyribo- and ribo-nucleotides, and combinations of bases, including uracil, adenine, thymine, cytosine, guanine, inosine, xanthine hypoxanthine, isocytosine, isoguanine, etc. As used herein, the term “nucleoside” includes nucleotides and nucleoside and nucleotide analogs, and modified nucleosides such as amino modified nucleosides. In addition, “nucleoside” includes non-naturally occurring analog structures. Thus, e.g. the individual units of a peptide nucleic acid, each containing a base, are referred to herein as a nucleoside.
  • The nucleic acid domain of a nucleic acid construct described herein is not limited by any particular sequence. Any number of oligonucleotide or polynucleotides useful for diagnostics, therapeutics and research can be used in the methods and compositions of the disclosure. Various sources of oligonucleotides and polynucleotides are available to one of skill in the art. For example, fragments of a genome may be isolated and the isolated polynucleotides modified in accordance with the disclosure to reduce the overall net anionic charge using phosphodiester and/or phosphothioate protecting groups or may be used as a source for extension of the oligonucleotide or polynucleotide using, for example, nucleic acid synthesis techniques known in the art.
  • The practice of phosphoramidite chemistry to prepare oligonucleotides is known from the published work of M. Caruthers and S. Beaucage and others. U.S. Pat. Nos. 4,458,066, 4,500,707, 5,132,418, 4,415,732, 4,668,777, 4,973,679, 5,278,302, 5,153,319, 5,218,103, 5,268,464, 5,000,307, 5,319,079, 4,659,774, 4,672,110, 4,517,338, 4,725,677 and Re. 34,069, each of which is herein incorporated by reference, describe methods of oligonucleotide synthesis. Additionally, the practice of phosphoramidite chemistry has been systematically reviewed by Beaucage and Iyer in Beaucage, S. L. and Iyer, R. P., Tetrahedron, 1992, 48, 2223-2311 and Beaucage, S. L. and Iyer, R. P., Tetrahedron, 1993, 49, 6123-6194, or references referred to therein, all of which are herein incorporated by reference.
  • Nucleic acid synthesizers are commercially available and their use is generally understood by persons of ordinary skill in the art as being effective in generating nearly any oligonucleotide of reasonable length which may be desired.
  • In practicing phosphoramidite chemistry useful 5′OH sugar blocking groups abbreviated to DMT in the figures, are trityl, momomethoxytrityl, dimethoxytrityl and trimethoxytrityl, especially dimethoxytrityl (DMTr). In practicing phosphoramidite chemistry useful phosphite activating groups, i.e., NR2, are dialkyl substituted nitrogen groups and nitrogen heterocycles. One approach includes the use of the di-isopropylamino activating group.
  • Various nucleoside units can be can be activated as amidites of the disclosure and incorporated in to the oligonucleotides or polynucleotides of the disclosure. These include deoxy nucleotides, i.e., wherein W in the above structures is H, ribonucleotides, in some embodiment W is F, i.e., wherein W is OH in the above structures, 2′-alkoxy nucleotides, i.e., wherein W is O-alkyl in the above structures, or substituted 2′-O-alkyl nucleotides, i.e., wherein W is substituted —O-alkyl in the above structures. 2′-O-alkyl nucleotides are described in U.S. Pat. No. 5,466,786, herein incorporated by reference. A particularly useful substituted 2′-O-alkyl group, the methoxyethoxy group, is described by Martin, P., Helv. Chim. Acta, 1995, 78, 486-504, also herein incorporated by reference.
  • Oligonucleotides can be synthesized by a Mermade −6 solid phase automated oligonucleotide synthesizer or any commonly available automated oligonucleotide synthesizer. Triester, phosphoramidite, or hydrogen phosphonate coupling chemistries described in, for example, M. Caruthers, Oligonucleotides: Antisense Inhibitors of Gene Expression., pp. 7-24, J. S. Cohen, ed. (CRC Press, Inc. Boca Raton, Fla., 1989) or Oligonucleotide synthesis, a practical approach, Ed. M. J. Gait, IRL Press, 1984; “Oligonucleotides and Analogues, A Practical Approach”, Ed. F. Eckstein, IRL Press, 1991, are employed by these synthesizers to provide the desired oligonucleotides. The Beaucage reagent, as described in, for example, Journal of American Chemical Society, 1990, 112, 1253-1255, or elemental sulfur, as described in Beaucage et al., Tetrahedron Letters, 1981, 22, 1859-1862, is used with phosphoramidite or hydrogen phosphonate chemistries to provide substituted phosphorothioate oligonucleotides. For example, the reagents comprising the protecting groups recited herein can be used in numerous applications where protection is desired. Such applications include, but are not limited to, both solid phase and solution phase, oligo- synthesis, polynucleotide synthesis and the like. The use of nucleoside and nucleotide analogs is also contemplated by this disclosure to provide oligonucleotide or oligonucleoside analogs bearing the protecting groups disclosed herein. Thus the terms nucleoside, nucleotide, deoxynucleoside and deoxynucleotide generally include analogs such as those described herein. These analogs are those molecules having some structural features in common with a naturally occurring nucleoside or nucleotide such that when incorporated into an oligonucleotide or oligonucleoside sequence, they allow hybridization with a naturally occurring oligonucleotide sequence in solution. Typically, these analogs are derived from naturally occurring nucleosides and nucleotides by replacing and/or modifying the base, the ribose or the phosphodiester moiety. The changes can be tailor made to stabilize or destabilize hybrid formation or enhance the specificity of hybridization with a complementary nucleic acid sequence as desired.
  • For instance, structural groups are optionally added to the ribose or base of a nucleoside for incorporation into an oligonucleotide, such as a methyl, propyl or allyl group at the 2′-0 position on the ribose, or a fluoro group which substitutes for the 2′-O group, or a bromo group on the ribonucleoside base. For use with phosphoramidite chemistry, various amidite reagents are commercially available, including 2′-deoxy amidites, 2′-O-methyl amidites and 2′-O-hydroxyl amidites. Any other means for such synthesis may also be employed. The actual synthesis of the oligonucleotides is well within the talents of those skilled in the art. It is also well known to use similar techniques to prepare other oligonucleotides such as the phosphorothioates, methyl phosphonates and alkylated derivatives. It is also well known to use similar techniques and commercially available modified amidites and controlled-pore glass (CPG) products such as biotin, Cy3, fluorescein, acridine or psoralen-modified amidites and/or CPG (available from Glen Research, Sterling Va.) to synthesize fluorescently labeled, biotinylated or other conjugated oligonucleotides.
  • Although the phosphotriester neutralizing/protecting groups described herein are useful for neutralizing the anionic charge of a nucleic acid domain, additional cationically charged moieties linked to a protected nucleic acid domain can be used to further facilitate uptake of oligonucleotide and polynucleotides. The recent discovery of several proteins which could efficiently pass through the plasma membrane of eukaryotic cells has led to the identification of a novel class of proteins from which peptide transduction domains have been derived.
  • A number of protein transduction domains/peptides are known in the art and have been demonstrated to facilitate uptake of heterologous molecules linked to the transdomain (e.g., cargo molecules). Such transduction domains facilitate uptake through a process referred to as macropinocytosis. Macropinocytosis is a nonselective form of endocytosis that all cells perform.
  • The best characterized of these proteins are the Drosophila homeoprotein antennapedia transcription protein (AntHD) (Joliot et al., New Biol. 3:1121-34, 1991; Joliot et al., Proc. Natl. Acad. Sci. USA, 88:1864-8, 1991; Le Roux et al., Proc. Natl. Acad. Sci. USA, 90:9120-4, 1993), the herpes simplex virus structural protein VP22 (Elliott and O'Hare, Cell 88:223-33, 1997), the HIV-1 transcriptional activator TAT protein (Green and Loewenstein, Cell 55:1179-1188, 1988; Frankel and Pabo, Cell 55:1189-1193, 1988), and more recently the cationic N-terminal domain of prion proteins. Not only can these proteins pass through the plasma membrane but the attachment of other proteins, such as the enzyme β-galactosidase, was sufficient to stimulate the cellular uptake of these complexes. Such chimeric proteins are present in a biologically active form within the cytoplasm and nucleus. Characterization of this process has shown that the uptake of these fusion polypeptides is rapid, often occurring within minutes, in a receptor independent fashion. Moreover, the transduction of these proteins does not appear to be affected by cell type and can efficiently transduce 100% of cells in culture with no apparent toxicity (Nagahara et al., Nat. Med. 4:1449-52, 1998). In addition to full-length proteins, protein transduction domains have also been used successfully to induce the intracellular uptake of DNA (Abu-Amer, supra), antisense oligonucleotides (Astriab-Fisher et al., Pharm. Res, 19:744-54, 2002), small molecules (Polyakov et al., Bioconjug. Chem. 11:762-71, 2000) and even inorganic 40 nanometer iron particles (Dodd et al., J. Immunol. Methods 256:89-105, 2001; Wunderbaldinger et al., Bioconjug. Chem. 13:264-8, 2002; Lewin et al., Nat. Biotechnol. 18:410-4, 2000; Josephson et al., Bioconjug., Chem. 10:186-91, 1999) suggesting that there is no apparent size restriction to this process. The effective transduction using transduction domains is, in part, limited by the overall molecular charge on the PTD-cargo construct.
  • The fusion of a protein transduction domain (PTD) with a heterologous molecule (e.g., a polynucleotide, small molecule, or protein) is sufficient to cause their transduction into a variety of different cells in a concentration-dependent manner. Moreover, this technique for protein delivery appears to circumvent many problems associated with DNA and drug based techniques.
  • PTDs are typically cationic in nature. These cationic protein transduction domains track into lipid raft endosomes carrying with them their linked cargo and release their cargo into the cytoplasm by disruption of the endosomal vesicle. Examples of PTDs include AntHD, TAT, VP22, cationic prion protein domains, poly-Arg, AGRKKRRQRRR (SEQ ID NO:15), YARKARRQARR (SEQ ID NO:16), YARAAARQARA (SEQ ID NO:17), YARAARRAARR (SEQ ID NO:18), YARAARRAARA (SEQ ID NO:19), YARRRRRRRRR (SEQ ID NO:20), YAAARRRRRRR (SEQ ID NO:21) and functional fragments and variants thereof. The disclosure provides, in one aspect, methods and compositions that combine the use of PTDs such as TAT and poly-Arg, with a charge neutralized nucleic acids. By charge neutralized is meant that the anionic charge of the nucleic acid (e.g., oligonucleotide or polynucleotide) is reduced, neutralized or more cationic than the same nucleic acid in the absence of a phosphodiester and/or phosphothioate protecting group or a phosphodiester and/or phosphothioate protecting group and a binding domain capable of neutralizing the anionic charge on a nucleic acid (i.e., the “cargo”) domain.
  • In general, the transduction domain of a nucleic acid construct of the disclosure can be nearly any synthetic or naturally-occurring amino acid sequence that can transduce or assist in the transduction of the fusion molecule. Typically, the transduction domain is cationically charged. For example, transduction can be achieved in accord with the disclosure by use of a nucleic acid construct including phosphodiester and/or phosphothioate protecting groups and a protein sequence such as an HIV TAT protein or fragment thereof that is linked at the N-terminal or C-terminal end to an oligonucleotide or polynucleotide comprising a phosphodiester and/or phosphothioate protecting group. In some aspects, the nucleic acid may comprise a phosphodiester and/or phosphothioate protecting group and may also comprise a nucleic acid binding domain (e.g., a DRBD). The transducing protein domain, for example, can be the Antennapedia homeodomain or the HSV VP22 sequence, the N-terminal fragment of a prion protein or suitable transducing fragments thereof such as those known in the art.
  • The type and size of the PTD will be guided by several parameters including the extent of transduction desired. PTDs will be capable of transducing at least about 20%, 25%, 50%, 75%, 80%, 90%, 95%, 98% 99% or 100% of the cells. Transduction efficiency, typically expressed as the percentage of transduced cells, can be determined by several conventional methods.
  • PTDs will manifest cell entry and exit rates (sometimes referred to as k1 and k2, respectively) that favor at least picomolar amounts of the fusion molecule in the cell. The entry and exit rates of the PTD and any cargo can be readily determined, or at least approximated, by standard kinetic analysis using detectably-labeled fusion molecules. Typically, the ratio of the entry rate to the exit rate will be in the range of between about 5 to about 100 up to about 1000.
  • In one aspect, a PTD useful in the methods and compositions of the disclosure comprise a peptide featuring substantial alpha-helicity. It has been discovered that transduction is optimized when the PTD exhibits significant alpha-helicity. In another embodiment, the PTD comprises a sequence containing basic amino acid residues that are substantially aligned along at least one face of the peptide. A PTD domain of the disclosure may be a naturally occurring peptide or a synthetic peptide.
  • In one embodiment of the disclosure, the PTD comprises an amino acid sequences comprising a strong alpha helical structure with arginine (Arg) residues down the helical cylinder. In yet another embodiment, the PTD domain comprises a peptide represented by the following general formula: B1—X1—X2—X3—B2—X4—X5—B3 (SEQ ID NO:1) wherein B1, B2, and B3 are each independently a basic amino acid, the same or different; and X1, X2, X3, X4 and X5 are each independently an alpha-helix enhancing amino acid, the same or different. In another embodiment, the PTD domain is represented by the following general formula: B1—X1—X2—B2—B3—X3—X4—B4 (SEQ ID NO:2) wherein B1, B2, B3, and B4 are each independently a basic amino acid, the same or different; and X1, X2, X3, and X4 are each independently an alpha-helix enhancing amino acid the same or different.
  • Additionally PTD domains comprise basic residues, e.g., lysine (Lys) or arginine (Arg), and further including at least one proline (Pro) residue sufficient to introduce “kinks” into the domain. Examples of such domains include the transduction domains of prions. For example, such a peptide comprises KKRPKPG (SEQ ID NO:3).
  • In one embodiment, the domain is a peptide represented by the following sequence: X—X—R—X—(P/X)—(B/X)—B—(P/X)—X—B—(B/X) (SEQ ID NO:4), wherein X is any alpha helical promoting residue such as alanine; P/X is either proline or X as previously defined; B is a basic amino acid residue, e.g., arginine (Arg) or lysine (Lys); R is arginine (Arg) and B/X is either B or X as defined above.
  • In another embodiment the PTD is cationic and consists of between 7 and 10 amino acids and has the formula K—X1—R—X2—X1 (SEQ ID NO:5) wherein X1 is R or K and X2 is any amino acid. An example of such a peptide comprises RKKRRQRRR (SEQ ID NO:6).
  • Additional transducing domains include a TAT fragment that comprises at least amino acids 49 to 56 of TAT up to about the full-length TAT sequence (see, e.g., SEQ ID NO:7). A TAT fragment may include one or more amino acid changes sufficient to increase the alpha-helicity of the fragment. In some instances, the amino acid changes introduced will involve adding a recognized alpha-helix enhancing amino acid. Alternatively, the amino acid changes will involve removing one or more amino acids from the TAT fragment that impede alpha helix formation or stability. In a more specific embodiment, the TAT fragment will include at least one amino acid substitution with an alpha-helix enhancing amino acid. Typically a TAT fragment or other PTD will be made by standard peptide synthesis techniques although recombinant DNA approaches may be used in some cases.
  • Additional transduction proteins (PTDs) that can be used in the nucleic acid constructs of the disclosure include the TAT fragment in which the TAT 49-56 sequence has been modified so that at least two basic amino acids in the sequence are substantially aligned along at least one face of the TAT fragment. Illustrative TAT fragments include at least one specified amino acid substitution in at least amino acids 49-56 of TAT which substitution aligns the basic amino acid residues of the 49-56 sequence along at least one face of the segment and typically the TAT 49-56 sequence.
  • Additional transduction proteins include the TAT fragment in which the TAT 49-56 sequence includes at least one substitution with an alpha-helix enhancing amino acid. In one embodiment, the substitution is selected so that at least two basic amino acid residues in the TAT fragment are substantially aligned along at least one face of that TAT fragment. In a more specific embodiment, the substitution is chosen so that at least two basic amino acid residues in the TAT 49-56 sequence are substantially aligned along at least one face of that sequence.
  • Also included are chimeric PTD domains. Such chimeric transducing proteins include parts of at least two different transducing proteins. For example, chimeric transducing proteins can be formed by fusing two different TAT fragments, e.g., one from HIV-1 and the other from HIV-2 or one from a prion protein and one from HIV.
  • PTDs can be linked or fused with any number of other molecules including an oligonucleotide or polynucleotide.
  • Alternatively, the nucleic acid construct or PTD can be bound to other molecular entities including nucleic acid binding domains, targeting moieties and the like. For example, two or more PTDs (e.g., 1-5, 2-4, typically 3) can be linked in series or separated by one or more other domains (e.g., a nucleic acid domain or peptide linkers). A nucleic acid binding domain can promote uptake of a fusion construct comprising a nucleic acid (including an oligonucleotide or polynucleotide comprising a protecting group) by reducing the anionic charge such that the cationic charge of the PTD domain is sufficient to transduce/traverse a cell's membrane. It will be understood that the PTD may be fused to an oligonucleotide or polynucleotide comprising a phosphodiester and/or phosphothioate protecting group and may further be linked to a nucleic acid binding domain. Exemplary RNA binding proteins (e.g., DRBD) include histone, RDE-4 protein, or protamine. Additional dsRNA binding proteins (and their Accession numbers in parenthesis) include: PKR (AAA36409, AAA61926, Q03963), TRBP (P97473, AAA36765), PACT (AAC25672, AAA49947, NP609646), Staufen (AAD17531, AAF98119, AAD17529, P25159), NFAR1 (AF167569), NFAR2 (AF167570, AAF31446, AAC71052, AAA19960, AAA19961, AAG22859), SPNR (AAK20832, AAF59924, A57284), RHA (CAA71668, AAC05725, AAF57297), NREBP (AAK07692, AAF23120, AAF54409, T33856), kanadaptin (AAK29177, AAB88191, AAF55582, NP499172, NP198700, BAB19354), HYL1 (NP563850), hyponastic leaves (CAC05659, BABOO641), ADAR1 (AAB97118, P55266, AAK16102, AAB51687, AF051275), ADAR2P78563, P51400, AAK17102, AAF63702), ADAR3 (AAF78094, AAB41862, AAF76894), TENR(XP059592, CAA59168), RNaseIII (AAF80558, AAF59169, Z81070Q02555/S55784, PO5797), and Dicer (BAA78691, AF408401, AAF56056, S44849, AAF03534, Q9884), RDE-4 (AY071926), FLJ20399 (NP060273, BAB26260), CG1434 (AAF48360, EAA12065, CAA21662), CG13139 (XP059208, XP143416, XP110450, AAF52926, EEA14824), DGCRK6 (BAB83032, XP110167) CG1800 (AAF57175, EAA08039), FLJ20036 (AAH22270, XP134159), MRP-L45 (BAB14234, XP129893), CG2109 (AAF52025), CG12493 (NP647927), CG10630 (AAF50777), CG17686 (AAD50502), T22A3.5 (CAB03384) and Accession number EAA14308.
  • As noted, two or more components of the constructs disclosed herein, e.g., a PTD, a nucleic acid binding domain, an oligonucleotide/polynucleotide domain, and peptide linkers, can be organized in nearly any fashion provided that the construct has the function for which it was intended (e.g., sufficiently cationic or having reduced anionic charge). The constructs can include fusion polypeptides or chimeric proteins comprising one or more PTDs linked either directly or indirectly linked to a oligonucleotide or polynucleotide domain (e.g., a therapeutic or diagnostic DNA, RNA, siRNA and the like). Each of the several domains may be directly linked or may be separated by a linker peptide. The domains may be presented in any order. Additionally, the fusion polypeptides may include tags, e.g., to facilitate identification and/or purification of the fusion polypeptide, such as a 6×HIS tag.
  • Peptide linkers that can be used in the fusion polypeptides and methods of the disclosure will typically comprise up to about 20 or 30 amino acids, commonly up to about 10 or 15 amino acids, and still more often from about 1 to 5 amino acids. The linker sequence is generally flexible so as not to hold the fusion molecule in a single rigid conformation. The linker sequence can be used, e.g., to space the PTD domain from the nucleic acid binding domain and/or nucleic acid domain. For example, the peptide linker sequence can be positioned between the protein transduction domain and the nucleic acid domain, e.g., to provide molecular flexibility. The length of the linker moiety is chosen to optimize the biological activity of the polypeptide comprising a PTD domain fusion construct and can be determined empirically without undue experimentation. The linker moiety should be long enough and flexible enough to allow a nucleic acid binding domain to freely interact with a nucleic acid or vice versa. Examples of linker moieties are -Gly-Gly-, GGGGS (SEQ ID NO:8), (GGGGS)N (SEQ ID NO:9), GKSSGSGSESKS (SEQ ID NO:10), GSTSGSGKSSEGKG (SEQ ID NO:11), GSTSGSGKSSEGSGSTKG (SEQ ID NO:12), GSTSGSGKPGSGEGSTKG (SEQ ID NO:13), or EGKSSGSGSESKEF (SEQ ID NO:14). Linking moieties are described, for example, in Huston et al., Proc. Nat'l Acad. Sci. 85:5879, 1988; Whitlow et al., Protein Engineering 6:989, 1993; and Newton et al., Biochemistry 35:545, 1996. Other suitable peptide linkers are those described in U.S. Pat. Nos. 4,751,180 and 4,935,233, which are hereby incorporated by reference.
  • The methods, compositions, and fusion polypeptides of the disclosure provide enhanced uptake and release of nucleic acid molecules by cells both in vitro and in vivo.
  • The term “therapeutic” is used in a generic sense and includes treating agents, prophylactic agents, and replacement agents. Examples of therapeutic molecules include, but are not limited to, cell cycle control agents; agents which inhibit cyclin proteins, such as antisense polynucleotides to the cyclin G1 and cyclin D1 genes; dsRNA that can be cleaved to provide siRNA molecules directed to specific growth factors such as, for example, epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), erythropoietin, G-CSF, GM-CSF, TGF-α, TGF-β, and fibroblast growth factor; cytokines, including, but not limited to, Interleukins 1 through 13 and tumor necrosis factors; anticoagulants, anti-platelet agents; TNF receptor domains etc.
  • Using such methods and compositions, various diseases and disorders can be treated. For example, growth of tumor cells can be inhibited, suppressed, or destroyed upon delivery of an anti-tumor siRNA.
  • Thus, it is to be understood that the disclosure is not to be limited to any particular transduction domain or oligonucleotide/polynucleotide. Any anionically charged nucleic acid (e.g., dsRNA, siRNA and the like) can be delivered using the methods of the disclosure.
  • The polypeptides used in the disclosure (e.g., with respect to particular domains of a fusion polypeptide or the full length fusion polypeptide) can comprise either the L-optical isomer or the D-optical isomer of amino acids or a combination of both. Polypeptides that can be used in the disclosure include modified sequences such as glycoproteins, retro-inverso polypeptides, D-amino acid modified polypeptides, and the like. A polypeptide includes naturally occurring proteins, as well as those which are recombinantly or synthetically synthesized. “Fragments” are a portion of a polypeptide. The term “fragment” refers to a portion of a polypeptide which exhibits at least one useful epitope or functional domain. The term “functional fragment” refers to fragments of a polypeptide that retain an activity of the polypeptide. For example, a functional fragment of a PTD includes a fragment which retains transduction activity. Biologically functional fragments, for example, can vary in size from a polypeptide fragment as small as an epitope capable of binding an antibody molecule, to a large polypeptide capable of participating in the characteristic induction or programming of phenotypic changes within a cell. An “epitope” is a region of a polypeptide capable of binding an immunoglobulin generated in response to contact with an antigen.
  • In some embodiments, retro-inverso peptides are used. “Retro-inverso” means an amino-carboxy inversion as well as enantiomeric change in one or more amino acids (i.e., levantory (L) to dextrorotary (D)). A polypeptide of the disclosure encompasses, for example, amino-carboxy inversions of the amino acid sequence, amino-carboxy inversions containing one or more D-amino acids, and non-inverted sequence containing one or more D-amino acids. Retro-inverso peptidomimetics that are stable and retain bioactivity can be devised as described by Brugidou et al. (Biochem. Biophys. Res. Comm. 214(2): 685-693, 1995) and Chorev et al. (Trends Biotechnol. 13(10): 438-445, 1995).
  • In another aspect, the disclosure provides a method of producing a fusion polypeptide comprising a PTD domain, a nucleic acid binding domain (e.g., DRBD) and a nucleic acid molecule by growing a host cell comprising a polynucleotide encoding the fusion polypeptide under conditions that allow expression of the polynucleotide, and recovering the fusion polypeptide. A polynucleotide encoding a fusion polypeptide of the disclosure can be operably linked to a promoter for expression in a prokaryotic or eukaryotic expression system. For example, such a polynucleotide can be incorporated in an expression vector to generate a fusion construct.
  • Accordingly, the disclosure also provides polynucleotides encoding a fusion protein construct of the disclosure. Such polynucleotides comprise sequences encoding one or more PTD domains, and/or a nucleic acid binding domain (e.g., DRBD). The polynucleotide may also encode linker domains that separate one or more of the PTDs and/or nucleic acid binding domains. In one aspect a fusion polypeptide comprising two or more PTD domains is produced and then linked to a charge reduced/protected oligonucleotide or polynucleotide.
  • Delivery of a polynucleotide of the disclosure can be achieved by introducing the polynucleotide into a cell using a variety of methods known to those of skill in the art. For example, a construct comprising such a polynucleotide can be delivered into a cell using a colloidal dispersion system. Alternatively, a polynucleotide construct can be incorporated (i.e., cloned) into an appropriate vector. For purposes of expression, the polynucleotide encoding a fusion polypeptide of the disclosure may be inserted into a recombinant expression vector. The term “recombinant expression vector” refers to a plasmid, virus, or other vehicle known in the art that has been manipulated by insertion or incorporation of a polynucleotide encoding a fusion polypeptide of the disclosure. The expression vector typically contains an origin of replication, a promoter, as well as specific genes that allow phenotypic selection of the transformed cells. Vectors suitable for such use include, but are not limited to, the T7-based expression vector for expression in bacteria (Rosenberg et al., Gene, 56:125, 1987), the pMSXND expression vector for expression in mammalian cells (Lee and Nathans, J. Biol. Chem., 263:3521, 1988), baculovirus-derived vectors for expression in insect cells, cauliflower mosaic virus, CaMV, and tobacco mosaic virus, TMV, for expression in plants.
  • Depending on the vector utilized, any of a number of suitable transcription and translation elements (regulatory sequences), including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, and the like may be used in the expression vector (see, e.g., Bitter et al., Methods in Enzymology, 153:516-544, 1987). These elements are well known to one of skill in the art.
  • The term “operably linked” and “operably associated” are used interchangeably herein to broadly refer to a chemical or physical coupling of two otherwise distinct domains that each have independent biological function. For example, operably linked refers to the functional linkage between a regulatory sequence and the polynucleotide regulated by the regulatory sequence. In another aspect, operably linked refers to the association of a nucleic acid domain and a transduction domain such that each domain retains its independent biological activity under appropriate conditions. Operably linked further refers to the link between encoded domains of the fusion polypeptides such that each domain is linked in-frame to give rise to the desired polypeptide sequence.
  • In yeast, a number of vectors containing constitutive or inducible promoters may be used (see, e.g., Current Protocols in Molecular Biology, Vol. 2, Ed. Ausubel et al., Greene Publish. Assoc. & Wiley Interscience, Ch. 13, 1988; Grant et al., “Expression and Secretion Vectors for Yeast,” in Methods in Enzymology, Eds. Wu & Grossman, Acad. Press, N.Y., Vol. 153, pp. 516-544, 1987; Glover, DNA Cloning, Vol. II, IRL Press, Wash., D.C., Ch. 3, 1986; “Bitter, Heterologous Gene Expression in Yeast,” Methods in Enzymology, Eds. Berger & Kimmel, Acad. Press, N.Y., Vol. 152, pp. 673-684, 1987; and The Molecular Biology of the Yeast Saccharomyces, Eds. Strathern et al., Cold Spring Harbor Press, Vols. I and II, 1982). A constitutive yeast promoter, such as ADH or LEU2, or an inducible promoter, such as GAL, may be used (“Cloning in Yeast,” Ch. 3, R. Rothstein In: DNA Cloning Vol. 11, A Practical Approach, Ed. DM Glover, IRL Press, Wash., D.C., 1986). Alternatively, vectors may be used which promote integration of foreign DNA sequences into the yeast chromosome.
  • An expression vector can be used to transform a host cell. By “transformation” is meant a permanent genetic change induced in a cell following incorporation of a polynucleotide exogenous to the cell. Where the cell is a mammalian cell, a permanent genetic change is generally achieved by introduction of the polynucleotide into the genome of the cell. By “transformed cell” or “recombinant host cell” is meant a cell into which (or into an ancestor of which) has been introduced, by means of molecular biology techniques, a polynucleotide encoding a fusion polypeptide of the disclosure. Transformation of a host cell may be carried out by conventional techniques as are known to those skilled in the art. Where the host is prokaryotic, such as E. coli, competent cells which are capable of polynucleotide uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCl2 method by procedures known in the art. Alternatively, MgCl2 or RbCl can be used. Transformation can also be performed after forming a protoplast of the host cell or by electroporation.
  • A fusion polypeptide of the disclosure can be produced by expression of polynucleotide encoding a fusion polypeptide in prokaryotes. These include, but are not limited to, microorganisms, such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA, or cosmid DNA expression vectors encoding a fusion polypeptide of the disclosure. The constructs can be expressed in E. coli in large scale. Purification from bacteria is simplified when the sequences include tags for one-step purification by nickel-chelate chromatography. Thus, a polynucleotide encoding a fusion polypeptide can also comprise a tag to simplify isolation of the fusion polypeptide. For example, a polyhistidine tag of, e.g., six histidine residues, can be incorporated at the amino terminal end of the fusion polypeptide. The polyhistidine tag allows convenient isolation of the protein in a single step by nickel-chelate chromatography. A fusion polypeptide of the disclosure can also be engineered to contain a cleavage site to aid in protein recovery the cleavage site may be part of a linker moiety as discussed above. A DNA sequence encoding a desired peptide linker can be inserted between, and in the same reading frame as, a polynucleotide encoding a PTD, or fragment thereof followed by a nucleic acid binding domain, the PTD may also be linked to a desired nucleic acid (e.g., dsRNA, DNA, siRNA, and the like), using any suitable conventional technique. For example, a chemically synthesized oligonucleotide encoding the linker can be ligated between two coding polynucleotides. In particular embodiments, a polynucleotide of the disclosure will encode a fusion polypeptide comprising from two to four separate domains (e.g., one or more PTD domain and one or more a nucleic acid domains) separated by linkers. In some embodiments, once purified, a fusion polypeptide comprising a plurality of PTDs is associated or linked with an oligonucleotide comprising a phosphodiester and/or phosphothioate protecting group or other anionic charge reducing group.
  • When the host cell is a eukaryotic cell, such methods of transfection of DNA as calcium phosphate co-precipitates, conventional mechanical procedures, such as microinjection, electroporation, insertion of a plasmid encased in liposomes, or virus vectors may be used. Eukaryotic cells can also be cotransfected with a polynucleotide encoding the PTD-fusion polypeptide of the disclosure, and a second polynucleotide molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene. Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the fusion polypeptide (see, e.g., Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982).
  • Eukaryotic systems, and typically mammalian expression systems, allow for proper post-translational modifications of expressed mammalian proteins to occur. Eukaryotic cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, phosphorylation, and advantageously secretion of the fusion product can be used as host cells for the expression of the PTD-fusion polypeptide of the disclosure. Such host cell lines may include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, Jurkat, HEK-293, and WI38.
  • For long-term, high-yield production of recombinant proteins, stable expression is used. Rather than using expression vectors that contain viral origins of replication, host cells can be transformed with the cDNA encoding a fusion polypeptide of the disclosure controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, and the like), and a selectable marker. The selectable marker in the recombinant plasmid confers selectivity (e.g., by cytotoxin resistance) and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci that, in turn, can be cloned and expanded into cell lines. For example, following the introduction of foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. A number of selection systems may be used, including, but not limited to, the herpes simplex virus thymidine kinase (Wigler et al., Cell, 11:223, 1977), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA, 48:2026, 1962), and adenine phosphoribosyltransferase (Lowy et al., Cell, 22:817, 1980) genes can be employed in tk-, hgprt- or aprt- cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate (Wigler et al., Proc. Natl. Acad. Sci. USA, 77:3567, 1980; O'Hare et al., Proc. Natl. Acad. Sci. USA, 8:1527, 1981); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci. USA, 78:2072, 1981; neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., J. Mol. Biol., 150:1, 1981); and hygro, which confers resistance to hygromycin genes (Santerre et al., Gene, 30:147, 1984). Additional selectable genes have been described, namely trpB, which allows cells to utilize indole in place of tryptophan; hisD, which allows cells to utilize histinol in place of histidine (Hartman & Mulligan, Proc. Natl. Acad. Sci. USA, 85:8047, 1988); and ODC (ornithine decarboxylase), which confers resistance to the ornithine decarboxylase inhibitor, 2-(difluoromethyl)-DL-ornithine, DFMO (McConlogue L., In: Current Communications in Molecular Biology, Cold Spring Harbor Laboratory, ed., 1987).
  • Techniques for the isolation and purification of either microbially or eukaryotically expressed PTD-fusion polypeptides of the disclosure may be by any conventional means, such as, for example, preparative chromatographic separations and immunological separations, such as those involving the use of monoclonal or polyclonal antibodies or antigen.
  • The fusion polypeptides of the disclosure are useful for the delivery of anionically charged nucleic acid molecules (e.g., dsRNA, siRNA, DNA, antisense, ribozymes and the like) for the treatment and/or diagnosis of a number of diseases and disorders. For example, the fusion polypeptides can be used in the treatment of cell proliferative disorders, wherein the protected oligo- or polynucleotide is reversibly modified such that it traverses a cell membrane alone or in associate with a PTD to target genes that induce cell proliferation. The PTD domain increases the overall net cationic charge or reduces the overall net anionic charge of the nucleic acid construct facilitating facilitates uptake by the cell. Thus, the constructs are useful for treatment of cells having cell proliferative disorders. Similarly, the constructs of the disclosure can be used to treat inflammatory diseases and disorders, infections, vascular disease and disorders and the like.
  • In one aspect, the construct of the disclosure may alternatively comprise, or in addition to, the PTD, a targeting domain. The targeting domain can be a receptor, receptor ligand or antibody useful for directing the construct to a particular cell type that expresses the cognate binding domain.
  • Typically a construct of the disclosure will be formulated with a pharmaceutically acceptable carrier, although the fusion polypeptide may be administered alone, as a pharmaceutical composition.
  • A pharmaceutical composition according to the disclosure can be prepared to include a fusion polypeptide of the disclosure, into a form suitable for administration to a subject using carriers, excipients, and additives or auxiliaries. Frequently used carriers or auxiliaries include magnesium carbonate, titanium dioxide, lactose, mannitol and other sugars, talc, milk protein, gelatin, starch, vitamins, cellulose and its derivatives, animal and vegetable oils, polyethylene glycols and solvents, such as sterile water, alcohols, glycerol, and polyhydric alcohols. Intravenous vehicles include fluid and nutrient replenishers. Preservatives include antimicrobial, anti-oxidants, chelating agents, and inert gases. Other pharmaceutically acceptable carriers include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like, as described, for instance, in Remington's Pharmaceutical Sciences, 15th ed., Easton: Mack Publishing Co., 1405-1412, 1461-1487 (1975), and The National Formulary XIV., 14th ed., Washington: American Pharmaceutical Association (1975), the contents of which are hereby incorporated by reference. The pH and exact concentration of the various components of the pharmaceutical composition are adjusted according to routine skills in the art. See Goodman and Gilman's, The Pharmacological Basis for Therapeutics (7th ed.).
  • The pharmaceutical compositions according to the disclosure may be administered locally or systemically. By “therapeutically effective dose” is meant the quantity of a fusion polypeptide according to the disclosure necessary to prevent, to cure, or at least partially arrest the symptoms of a disease or disorder (e.g., to inhibit cellular proliferation). Amounts effective for this use will, of course, depend on the severity of the disease and the weight and general state of the subject. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of the pharmaceutical composition, and animal models may be used to determine effective dosages for treatment of particular disorders. Various considerations are described, e.g., in Langer, Science, 249: 1527, (1990); Gilman et al. (eds.) (1990), each of which is herein incorporated by reference.
  • As used herein, “administering a therapeutically effective amount” is intended to include methods of giving or applying a pharmaceutical composition of the disclosure to a subject that allow the composition to perform its intended therapeutic function. The therapeutically effective amounts will vary according to factors, such as the degree of infection in a subject, the age, sex, and weight of the individual. Dosage regima can be adjusted to provide the optimum therapeutic response. For example, several divided doses can be administered daily or the dose can be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • The pharmaceutical composition can be administered in a convenient manner, such as by injection (e.g., subcutaneous, intravenous, and the like), oral administration, inhalation, transdermal application, or rectal administration. Depending on the route of administration, the pharmaceutical composition can be coated with a material to protect the pharmaceutical composition from the action of enzymes, acids, and other natural conditions that may inactivate the pharmaceutical composition. The pharmaceutical composition can also be administered parenterally or intraperitoneally. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof, and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. The composition will typically be sterile and fluid to the extent that easy syringability exists. Typically the composition will be stable under the conditions of manufacture and storage and preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size, in the case of dispersion, and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, isotonic agents, for example, sugars, polyalcohols, such as mannitol, sorbitol, or sodium chloride are used in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the pharmaceutical composition in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the pharmaceutical composition into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • The pharmaceutical composition can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The pharmaceutical composition and other ingredients can also be enclosed in a hard or soft-shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the pharmaceutical composition can be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 1% by weight of active compound. The percentage of the compositions and preparations can, of course, be varied and can conveniently be between about 5% to about 80% of the weight of the unit.
  • The tablets, troches, pills, capsules, and the like can also contain the following: a binder, such as gum gragacanth, acacia, corn starch, or gelatin; excipients such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid, and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin, or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring. When the dosage unit form is a capsule, it can contain, in addition to materials of the above type, a liquid carrier. Various other materials can be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules can be coated with shellac, sugar, or both. A syrup or elixir can contain the agent, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye, and flavoring, such as cherry or orange flavor. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the pharmaceutical composition can be incorporated into sustained-release preparations and formulations.
  • Thus, a “pharmaceutically acceptable carrier” is intended to include solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the pharmaceutical composition, use thereof in the therapeutic compositions and methods of treatment is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. “Dosage unit form” as used herein, refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of pharmaceutical composition is calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the disclosure are related to the characteristics of the pharmaceutical composition and the particular therapeutic effect to be achieve.
  • The principal pharmaceutical composition is compounded for convenient and effective administration in effective amounts with a suitable pharmaceutically acceptable carrier in an acceptable dosage unit. In the case of compositions containing supplementary active ingredients, the dosages are determined by reference to the usual dose and manner of administration of the said ingredients.
  • EXAMPLES
  • RNB Synthesis example.
  • Figure US20090093425A1-20090409-C00030
  • Phenyl chloroformate is added to Guanidine hydrochloride under basic conditions. Selective ring opening of thirane is accomplished under lewis acidic conditions to provide the free thiol. Thiolated material is linked to betamercaptoethanol via a disulfide linkage to provide a free alcohol. Reaction of the free alcohol with tetraisopropylphosphonamidic chloride provides the diamidite capable of reaction with the free 3′OH of any nucleoside amidite precursor. Oligonucleotides were synthesized using techniques known in the art as described herein.
  • RNN Phosphoramidite protection.
  • Figure US20090093425A1-20090409-C00031
  • Ribonucleic Base RNB protecting groups.
  • Figure US20090093425A1-20090409-C00032
  • Reactions were carried out under an argon atmosphere. Glassware was cleaned overnight in a KOH/EtOH base bath, rinsed with MeOH and flame dried under vacuum before use in all anhydrous systems. Reactions were run with solvents that were either purchased sure-sealed over molecular sieves or were distilled using protocols listed in Purification of Laboratory Chemicals 4th ed. and stored over sieves.
  • Tetrahydrofuran (THF) was distilled from sodium metal and benzophenone, triethyl amine (Et3N), di-isopropyl amine (DIEA) and pyridine (py) were distilled from sodium metal. Dichloromethane (CH2Cl2), methanol (MeOH) and toluene were distilled from calcium hydride. All other solvents and reagents were purchased from Fisher Chemical Co., Aldrich Chemical Co., EMD or Acros Organics and used without further purification. Reactions were cooled to −78° C. via dry ice-acetone baths.
  • Flash column chromatography was performed using Merck grade 60 silica gel (230-400 mesh) and TLC analysis was carried out using Merck 60F-254 pre-coated silica sheets. Visualization of TLC plates was achieved using ultraviolet light, p-anisaldehyde in ethanol with sulfuric acid, polyphosphomolybdic acid and cerium sulfate in EtOH with H2SO4, ninhydrin in EtOH with H2SO4, potassium permanganate or iodine. Solvent removal was effected by Büchi rotary evaporator equipped with a dry ice isopropanol cold finger trap, and a H2O aspirator was used to concentrate in vacuo. Samples were further dried under reduced pressure on a high vacuum line over P2O5 and KOH. 1H NMR spectra were taken on a Varian Unity Inova 400 in CDCl3 or d6Benzene at ambient temperature unless otherwise noted. Mestre-C was used to visualize and measure J couplings. 1H chemical shifts in CDCl3 were reported in ppm; (δ units) downfield from tetramethylsilane. 1H NMR splitting patterns are designated as a singlet (s), doublet (d), triplet (t) or quartet (q). All first order splitting patterns were assigned based on the appearance of the multiplet as interpreted by the program. Splitting patterns that could not be easily interpreted are designated as multiplet (m) or (br). In cases where broad or multiplet splitting patterns are clearly centered on one peak the chemical shift for that peak was reported instead of a range. The solvent peak at 2.49 was used as an internal reference in DMSO-d6, and the solvent peak at 3.30 was used as an internal reference in CD3OD. Solvent peaks were used as internal references for all 13C NMR. NMR spectral data is tabulated as follows: chemical shift, multiplicity, coupling constant and number of protons. 31p NMR chemical shifts were measured relative to a phosphoric acid standard and spectra were acquired in d6 benzene unless otherwise noted. NMR 13C and 1H data were not reported when the product was a racemic mixture. Mass spectroscopy was obtained at the HHMI Analytical Instrumentation Center (AIC) of the University of California San Diego.
  • Oligonucleotides of 17-21 nt in length were synthesized on an MerMade 6 automated DNA/RNA synthesizer. Glen Research Q CPG support was used with ethylthioltetrazole as the coupling reagent during 3 coupling steps of 5 minutes each. Phosphine was oxidized to phosphate by the standard iodine method and capping was performed with phenoxyacetic anhydride. All amidites and materials used on the MerMade 6 synthesizer were either synthesized or purchased from EMD, ChemGenes or Glen Research.
  • Figure US20090093425A1-20090409-C00033
  • Examples of alcohol synthesis (Scheme II). Modified amidites were synthesized by preparing the free alcohol or thiol in the configurations shown below.
  • Figure US20090093425A1-20090409-C00034
  • 2-((hydroxymethyl)disulfanyl)ethyl acetate
  • VI019: 2 hydroxyethyl disulfide (1.2 ml, 10 mmol), Imidazole (1.23 g, 10 mmol) and DMF were placed in a flask equipped with a stir bar at room temperature. Acetylchloride (639.4 g, 9 mmol) was dissolved in DMF and added over 2 h by syringe pump to the stirring solution. After 6 h the solution was dissolved in EtOAc and extracted with saturated sodium bicarbonate, brine and dried over sodium sulfate. Ethyl acetate was removed and the resulting oil was fractionated over a silica column. Fractions containing product were pooled and solvent was removed to give 635 mg of a colorless oil at 36%.
  • Figure US20090093425A1-20090409-C00035
  • ((2-(tert-butyldimethylsilyloxy)ethyl)disulfanyl)methanol
  • VI139: 2 hydroxyethyl disulfide (19.17 g. 160 mmol), Imidazole (20.7 g, 304 mmol) and DMF (100 ml) were placed in a flask equipped with a stir bar at room temperature. TBSCl (38.9 ml, 152 mmol) was dissolved in DMF (60 ml) and added over 2 h by syringe pump to the stirring solution. After 15 h the solution was dissolved in EtOAc and extracted with sodium bicarbonate, brine and dried over sodium sulfate. Evaporation gave an oil that was applied to a silica column. Fractions containing product were pooled and evaporated to give 18.8 g of a light amber oil at 31%. (M+H)=268.74. 1H NMR (500 MHz, Solvent) δ ppm 3.92-3.80 (m, 4H), 2.90-2.75 (m, 4H), 2.50 (s, 1H), 0.92-0.82 (m, 9H), 0.05 (m, 6H).
  • Figure US20090093425A1-20090409-C00036
  • 2-((2-tert-butyldiphenylsilyloxy)ethyl)disulfanyl)ethanol
  • VI063: 2 hydroxyethyl disulfide (2.4 ml, 20 mmol), Imidazole 5.8 g, 85.5 mmol) and 20 ml DMF were placed in a flask equipped with a stir bar at room temperature. TBDPSCl (4.9 ml, 19 mmol) was dissolved in DMF and added over 2 h by syringe pump to the stirring solution. After 15 h the solution was dissolved in EtOAc and extracted. Silica chromatography yielded 1.79 g of a light amber oil at 24%. 1H NMR (500 MHz, CDCl3) δ ppm 7.82-7.64 (m, 1H), 7.50-7.38 (m, 1H), 5.32-5.30 (m, 1H), 3.99-3.91 (m, 1H), 3.91-3.84 (m, 1H), 2.93-2.87 (m, 1H), 2.81-2.77 (m, 1H), 2.34-2.31 (m, 1H), 1.13-1.08 (m, 1H).
  • Figure US20090093425A1-20090409-C00037
  • S-2-hydroxyethyl 2,2-dimethylpropanethioate
  • VI183: Mercaptoethanol (10 ml, 142.6 mmol) and Triethyl amine (19.9 ml, 142.6 mmol) were dissolved in dichloromethane 275 ml and cooled to −78° C. Pivaloyl chloride (17.56 ml, 142.6 mmol) was added dropwise to the stirring solution. The reaction was allowed to proceed for one hour at −78° C. then it was warmed to rt and stirred for an additional 1 h before quenching with 150 ml water. The aqueous was washed (3×100 ml DCM) and dried with magnesium sulfate. Quantitative conversion was observed by TLC crude was diluted with Hexanes and run over a short silica column to give 19.6 g of colorless oil at 85%. 1H NMR (400 MHz, CDCl3) δppm 3.61 (dd, J=11.79 Hz, 2H), 3.05 (t, J=5.46 Hz, 1H), 2.95 (td, J=12.60, 3.42 Hz, 2H), 1.14 (d, J=6.18 Hz, 9H). 13C NMR (101 MHz, CDCl3 δ ppm (207.2, 61.4, 46.3, 31.0, 27.1 ppm.
  • Figure US20090093425A1-20090409-C00038
  • 3-(hydroxymethyl)dihydrothiophen-2(3H)-one
  • VI269: Thiobutyrolactone (5.1 g, 50 mmol) in dry THF was added dropwise to a stirred solution of lithium diisopropylamide. (Diisopropylamine (5.1 mg, 50.5 mmol) and n-butyllithium in hexane (31.3 ml, 50 mmol) at −78° C.). The resulting solution was stirred for 10 minutes at which time formaldehyde (31.3 g) carried in a stream of argon was added (Formaldehyde was formed by heating p-formaldehyde to 150 C.) The reaction was allowed to proceed for 2.5 h at −78 C. The formaldehyde stream was removed and reaction was allowed to proceed for an additional 30 m. The reaction was quenched by the addition of (˜100 ml) 1M HCl at −78 C then allowed to warm to rt and filtered through a bed of celite. The filtrate was extracted with ethyl acetate 5×100 ml and the combined organic layers were dried (Na2SO4) and concentrated to an oil. The oil was chromatographically isolated to give 1.1 g of isolate at 17%.
  • Figure US20090093425A1-20090409-C00039
  • N-((2-hydroxethylthio)methyl)acetamide
  • IV185: Acetamide (890 mg, 10 mmol) was dissolved in TFA (10 ml) at rt. Mercaptoethanol (700 mg, 10 mmol) was added dropwise to the stirring solution. The reaction was allowed to proceed for 30 m. TFA was evaporated and the resulting oil was recrystallized from aqueous 2 propanol.
  • Examples of Diamidite formation:
  • Figure US20090093425A1-20090409-C00040
  • 1-butoxy-N,N,N′,N′-tetraisopropylphosphinediamine
  • VI173: n-butanol (190 ul, 11.0 mmol), triethylamine (418 ul, 3 mmol), and benzene were placed in a flask equipped with a stir bar at room temperature. Chlorophosphine (292 mg, 1.1 mmol) was added to the stirring solution and the reaction was allowed to progress for 2 h. Solvent was removed from the resulting mixture and the oil was applied directly to a triethylamine pretreated column. Fractions containing product were evaporated to give 311 mg oil at 64%.
  • Figure US20090093425A1-20090409-C00041
  • 1-(3,3 dimethylbutoxy)-N,N,N′,N′-tetraisopropylphosphirediamine
  • VI257: Tetraisopropyl phosphoramidic chloride (2.0 g, 7.5 mmol) was transferred under a stream of argon to a flame dried reaction vessel equipped with a stir bar at room temperature. Triethylamine (1.05 ml, 7.5 mmol) and benzene (7 ml) were added via syringe. The reaction was started by addition of 2′2′ dimethyl butanol (700 mg, 6.84 mmol) by syringe. The reaction was monitored for complete conversion by TLC over the course of 2.5 h. Solvent was removed from the mixture and the resulting oil was applied directly to a TEA pretreated column. Solvent was removed from fractions containing product and the resulting oil was co-evaporated 2× with toluene and lyophilized from benzene overnight to give 1.6 g colorless solid at 71%.
  • Figure US20090093425A1-20090409-C00042
  • S-2-(bis(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • VI253 SPTE: Tetraisopropyl phosphoramidic chloride (5.0 g, 18.81 mmol) was transferred under a stream of argon to a flame dried reaction vessel equipped with a stir bar at room temperature. Triethylamine (2.6 ml, 18.8 mmol) and benzene (20 ml) were added via syringe. The reaction was started by addition of the thioester VI183 (2.77 g, 17.1 mmol) by syringe. The reaction was monitored for complete conversion by TLC over the course of 2.5 h. Solvent was removed from the mixture and the resulting oil was applied directly to a TEA pretreated column. Solvent was removed from fractions containing product and the resulting oil was co-evaporated 2× with toluene and lyophilized from benzene overnight to give 6.08 g colorless solid at 91%. 31P NMR (162 MHz, d6-Benzene δ ppm 123.5 ppm. 13C NMR (101 MHz, CDCl3 δ ppm 205.2, 63.6, 63.4, 44.7, 44.6, 30.8, 30.7, 27.4, 24.7, 24.6, 24.0, 24.0 ppm. 1H NMR (400 MHz, Solvent δ ppm 3.70 (dd, J=13.46 Hz, 2H), 3.48 (dq, J=13.44 Hz, 4H), 3.14 (t, J=6.34 Hz, 2H), 1.25-1.11 (m, 33H).
  • Figure US20090093425A1-20090409-C00043
  • N,N,N′,N′-tetraisopropyl-11,11-dimethyl-10,10 diphenyl-2,9-dioxa-5,6-dithia-1-phospha-10-siladodecane-1,1-diamine
  • VI167: Bis (diisopropylamino) chlorophosphine (2 g, 7.5 mmol) IV139 (3.02 g, 7.7 mmol) and diisopropyl amine (2 ml, 15 mmol) were reacted in 10 ml benzene. After chromatography 4.09 of oil was collected at 86%.
  • Figure US20090093425A1-20090409-C00044
  • 4-(diisopropylamino)-isopropyl-2-methyl-5-oxa-8,9-dithia-3-aza-4-phosphaundecan-11-yl benzoate
  • VI171: 2 hydroxyethyl disulfide (910 ml, 7.6 mmol), Triethylamine (3.2 ml, 22.8 mmol), and benzene were placed in a flask equipped with a stir bar at room temperature. To the stirring solution was added benzoyl chloride (970 ul, 8.36 mmol). The reaction was monitored for complete conversion of disulfide by TLC. The reaction was diluted with 10 ml ether and filtered into a fresh reaction vessel under anhydrous conditions. Chlorophosphine (1 g, 3.76 mmol) was added dry under a stream of argon to the stirring solution and the reaction allowed to progress for an additional 2 h. Solvent was removed and the resulting oil was applied directly to a TEA pretreated column. Solvent was removed from collected fractions to give 648 mg of a colorless oil at 35%.
  • Error! Objects cannot be created from editing field codes.
  • VI147: Nucleoside (132.2 mg, 0.2 mmol) and diisopropylethylamine amine (105 ul, 0.6 mmol) were diluted with DCM. This solution was slowly added to a dilution of phosphorus trichloride 2M in dichloromethane (100 ul) dropwise. Flask was equipped with a vent needle. Completion conversion of starting material was observed at by TLC. Solvent and excess phosphorus trichloride were removed from the reaction mixture by evaporation. The reaction was resuspended in anhydrous ether and filtered to remove amine salts and unreacted reacted phosphine. The ether was evaporated and the resulting crude solid was taken on to the next step of the reaction series directly due to issues of spontaneous decomposition.
  • Figure US20090093425A1-20090409-C00045
  • 1-((2R,4R,5R)-5-(bis(4 methoxyphenyl)(phenyl)methoxy)methyl)-4-(bis(diisopropylamino)phosphinooxy)tetrahydrofuran-2-yl)-5-methylpyrimidine-2,4(1H,3H)-dione
  • VI179: Nucleoside (1.53 g, 2.81 mmol) and diisopropylethylamine amine (11.0 ml, 5.63 mmol) were diluted with 10 ml dichloromethane. This solution was slowly added to a dilution of chloroamidite (997 mg, 3.75 mmol) in 10 ml dichloromethane in a dropwise manner. The reaction was allowed to proceed for 30 min at which point a vent needle was placed in the flask to allow HCl to escape with the argon flow. The reaction vessel was heated to with a heat gun and the vent was removed. Completion conversion of starting material was observed at by TLC. Solvent was removed from the reaction mixture the reaction was resuspended in anhydrous ether and filtered to remove amine salts and unreacted chloroamidite. At 4 h the reaction was quenched with 1 ml TEA and evaporated to give an oil that was loaded directly onto a 1% TEA pretreated silica plug. Fractions containing product were pooled and evaporated to give 2.86 g of a pale yellow solid at 85%. 31p NMR (162 MHz, d6-Benzene) δ ppm 115.1.
  • Example of nucleoside activation as an phosphotriester amidite for solid phase synthesis: Nucleoside amidites were prepared in a manner suitable for the Caruthers method of solid phase oligonucleotide synthesis. Nucleoside and activator are added to a flame dried vial. Phosphoramidite is added to the stirring solution reaction was allowed to proceed for a minimum of 3 h. Solvent was removed from the reactions and the resulting foam or oil was applied directly to a triethylamine pretreated silica column in running buffer. Fractions containing only product were pooled and the solvent was removed. The resulting foam was dissolved in acetonitrile and syringe filtered (0.45 um). Acetonitrile was removed and the resulting foam was dissolved in benzene and lyophilized to remove all traces of triethyl amine and residual water. Samples were sealed in glass containers under argon and stored at −20° C. prior to use on the automated oligonucleoside synthesizer.
  • Figure US20090093425A1-20090409-C00046
  • In some aspect, phenooxyacetal base protection was used during synthesis. In others, a hindered base for cleavage form the solid support, which did not use a photolabile linker was used. One advantage of the disclosure is the ability to use either or both of DNA and RNA nucleotides.
  • Synthetic route with 2° F. and 2′2′ dimethyl butanol phosphate protection:
  • Figure US20090093425A1-20090409-C00047
  • (2R,3S,4S,5R)-5-(4-acetamido-2-oxopyrimidin-1(2H)-yl)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-4-fluorotetrahydrofuran-3-yl 3,3-dimethybutly diisopropylphosphoranidite
  • VI261: Protected cytosine (840 mg, 1.43 mmol) and 2′2′dimethyl phosphordiamidite VI257 (498.5 mg, 1.5 mmol) were dissolved in 5.7 ml dichloromethane. Ethylthiotetrazole 0.45M in acetonitrile (5.7 ml) was added and the reaction was found to be complete in 2.5 h. Chromatography and filtration resulted in 800 mg lyophilate at 68.4%.
  • Figure US20090093425A1-20090409-C00048
  • (2R,3S,4S,5R)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluorotetrahydrofuran-3-yl 3,3-dimethylbutyl diisopropylphosphoramidite
  • VI261: Protected uridine (782 mg, 1.43 mmol) and 2′2′dimethyl phosphordiamidite VI257 (498.5 mg, 1.5 mmol) were dissolved in 5.7 ml dichloromethane. Ethylthiotetrazole 0.45M in acetonitrile (5.7 ml) was added and the reaction was found to be complete in 2.5 h. Chromatography and filtration resulted in 860 mg lyophilate at 77.4%.
  • Synthetic route to 2° F. methylphosphoramidites:
  • Figure US20090093425A1-20090409-C00049
  • Commercially available, (Carbosynth, R.I. Chemical) free 3′OH nucleobases were reacted directly with the chlorophosphine under basic conditions in benzene.
  • Figure US20090093425A1-20090409-C00050
  • (2R,3S,4S,5R)-5-(4-acetamido-2-oxopyrimidin-1(2H)-yl 2-(bis(4 methoxyphenyl)(phenyl)methoxy)methyl)-4-fluoratetrahydrofuran-3-yl methyl diisopropylphosphoranidte
  • VI245, 237: Protected cytosine nucleoside (1.0 g, 1.7 mmol) and DIEA (1.42 ml, 8.2 mmol) were added to a flame dried vial. To the stirring solution was added phosphonamidic chloride (1.34 g, 6.79 mmol) and the reaction was allowed to proceed for 3 h. The reaction was concentrated to an oil and applied directly onto a TEA pretreated column. Isolation was achieved by chromatography DCM:Ethyl acetate:TEA 45:45:10. Fractions containing product were evaporated followed by transfer/filtration through a 0.45 um filter in acetonitrile. Lyophilization from benzene gave 1.02 g lyophilate at 80.4%.
  • Figure US20090093425A1-20090409-C00051
  • (2R,3S,4S,5R)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluorotetrahydrofuran-3-yl methyl diisopropylphosphoramidite
  • VI251: Protected Uridine nucleoside (2.74 g, 5.0 mmol) and DIEA (4.2 ml, 24 mmol) were added to a flame dried vial. To the stirring solution was added phosphonamidic chloride (3.94 g, 20.0 mmol) and the reaction was allowed to proceed for 3 h. The reaction was concentrated to an oil and applied directly onto a TEA pretreated column. Isolation was achieved by chromatography Hexane: DCM:Ethyl acetate:TEA 45:45:45:10. Fractions containing product were evaporated followed by transfer/filtration through a 0.45 um filter in acetonitrile. Lyophilization from benzene gave 2.46 g lyophilate at 84%.
  • Synthetic Route with SPTE biolabile protecting groups:
  • Figure US20090093425A1-20090409-C00052
  • Figure US20090093425A1-20090409-C00053
  • 2-(((2R,3R,5R)-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-5-(5-methyl-2,4-dioxo-3,4-dihdropyrimidin-1(2H)-yl)tetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • VI247: Protected Nucleoside (2.07 g, 3.08 mmol) and ethylthiotetrazole (15.2 ml) were diluted with DCM. To the stirring solution was added the phosphorodiamidite VI253 (1.57 g, 4.0 mmol) and the reaction was allowed to proceed for 30 m. The reaction was quenched by pouring the crude into a saturated solution of sodium bicarbonate and washing with brine washed 3×10 ml. The organic was dried over NaSO4 and evaporated. Isolation was achieved by chromatography Hexane:DCM:EthylAcetate:TEA 40:40:20:7.5. Fractions containing product were evaporated followed by transfer/filtration through a 0.45 um filter in benzene and lyophilization from benzene overnight to ensure complete removal of water. 2.74 g lyophilate was recovered at 86%. 31p NMR (162 MHz, d6-Benzene) δ ppm 148.7, 148.2.
  • Figure US20090093425A1-20090409-C00054
  • S-2-(((2R,3R,5R)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-5-(6-(2-phenoxyacetamido)-9H-purin-9-yl)tetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • VI267: 5′ DMT phenoxyacyldeoxyadenosine (3.92 g, 5.7 mmol) and ethylthiotetrazole activator (22.8 ml, 5.7 mmoles) were added to a flame dried vial and diluted in dichlomethane (22.8 ml). To the stirring solution was added the phosphorodiamidite VI253 (2.36 g, 6.0 mmol) and the reaction was allowed to proceed for 3 h. Solvent was removed from the reaction vessel and loaded directly on a TEA pretreated column. Fractions containing product Rf=0.5 were pooled and evaporated to an oil. The oil was suspended in acetonitrile, filtered with a 0.45 um syringe filter, evaporated to a foam, dissolved in benzene and lyophilized to give 3.98 g colorless lyophylate at 71%.
  • 2′OTBS Phosphine SPTE protected nucleosides
  • Figure US20090093425A1-20090409-C00055
  • S-2-(((2R,3S,4S,5R)-2-(bis(4-methoxyphenyl(phenyl)methoxy)methyl)-4-(tert-butyldimethylsilyloxy)-5-(6-(2-phenoxyacetamido)-9H-purin-9-yl))tetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • VI199: Protected nucleoside (2.33 g, 2.85 mmol) and hydrotetrazolide (488 mg, 2.85 mmol) were diluted with DCM. To the stirring solution was added the phosphorodiamidite (1.18 g, 3.0 mmol) and the reaction was allowed to proceed for 30 m. The reaction was quenched by pouring the crude into a saturated solution of sodium bicarbonate and washing with brine washed 3×10 ml. The organic was dried over NaSO4 and evaporated. Isolation was achieved by chromatography DCM:EthylAcetate:TEA 45:45:10. Fractions containing product were evaporated followed by transfer/filtration through a 0.2 um filter in benzene and lyophilization from benzene overnight to ensure complete removal of water. 2.1 g lyophilate was recovered at 67%. 31P NMR (162 MHz, d6-Benzene) δ ppm 150.34, 148.79.
  • Figure US20090093425A1-20090409-C00056
  • S-2-(((2R,3S,4S,5R)-2-(bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-4-(tert-butyldimethylsilyloxy)-5-(6-oxo-2-(2-phenoxyacetamido)-1H-purin-9(6H)-yl)tetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethypropanethioate
  • VI201: Protected nucleoside (2.38 g, 2.85 mmol) and hydrotetrazolide (448 mg, 2.85 mmol) were diluted with DCM. To the stirring solution was added the phosphorodiamidite VI253 (1.18 g, 3 mmol) and the reaction was allowed to proceed for 30 m. The reaction was quenched by pouring the crude into a saturated solution of sodium bicarbonate and washing with brine washed 3×10 ml. The organic was dried over NaSO4 and evaporated. Isolation was achieved by chromatography DCM:EthylAcetate:TEA 45:45:10. Fractions containing product were evaporated followed by transfer/filtration through a 0.45 um filter in benzene and lyophilization from benzene overnight to ensure complete removal of water. Recovered 815 mg lyophilate recovered at 25.4%. 31p NMR (162 MHz, d6-Benzene) δ ppm 149.22, 149.15.
  • Figure US20090093425A1-20090409-C00057
  • S-2-(((2R,3S,4S,5R)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-4-(tert-butyldimethylsilyloxy)-5-(2-oxo-4-(2-phenoxyacetamido)pyrimidin-1(2H)-yl)tetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • VI207: Protected cytosine (3.77 g, 4.75 mmol) and SPTE phosphordiamidite VI253 (1.96 g, 5.0 mmol) were dissolved in 19 ml dichloromethane. Ethylthiotetrazole 0.45M in acetonitrile (19 ml) was added and the reaction was found to be complete in 2.5 h. Chromatography and filtration resulted in 4.3glyophilate at 83.4%. 31p NMR (162 MHz, d6-Benzene) δ ppm 150.5, 147.6.
  • Figure US20090093425A1-20090409-C00058
  • S-2-(((2R,3S,4S,5R)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-4-(tert-butyldimethylsilyloxy)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)tetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • VI205: Protected uridine (3.42 g, 5.18 mmol) and SPTE phosphordiamidite VI253 (2.1399, 545 mmol) were dissolved in 20.7 ml dichloromethane. Ethylthiotetrazole 0.45M in acetonitrile (20.7) was added and the reaction was found to be complete in 2.5 h. Chromatography and filtration resulted in 4.7 g lyophilate at 95%. 31P NMR (162 MHz, d6-Benzene) δ ppm 149.9, 149.1.
  • Figure US20090093425A1-20090409-C00059
  • S-2-(((2R,3S,4S,5R)-5-(4-acetamido-2-oxopyrimidin-1(2H)-yl)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-4-fluorotetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • 2′ F. nucleosides containing SPTE phosphine protecting groups:
  • VI239: Protected cytosine (2.0 g, 3.39 mmol) and SPTE phosphordiamidite (14.02 g, 3.57 mmol) were dissolved in 13.6 ml dichloromethane. Ethylthiotetrazole 0.45M in acetonitrile (13.6 ml) was added and the reaction was found to be complete in 2.5 h. Chromatography and filtration resulted in 2.22 g lyophilate at 74%. 31P NMR (162 MHz, d6-Benzene) δ ppm 150.3, 150.2 ppm.
  • Figure US20090093425A1-20090409-C00060
  • S-2-(((2R,3S,4S,5R)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-4-fluorotetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • VI249: Protected uridine (2.08 g, 3.9 mmol) and SPTE phosphordiamidite (15.7 g, 4.0 mmol) were dissolved in 15.2 ml dichloromethane. Ethylthiotetrazole 0.45M in acetonitrile (15.2 ml) was added and the reaction was found to be complete in 2.5 h. Chromatography and filtration resulted in 2.6 g lyophilate at 82%.
  • 2′OMe modified oligonucleosides containing an SPTE phosphate protecting group
  • Figure US20090093425A1-20090409-C00061
  • S-2-(((2R,3S,4S,5R)-2-((bis(4-methoxyphenyl)(phenyl)methoxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-methoxytetrahydrofuran-3-yloxy)(diisopropylamino)phosphinooxy)ethyl 2,2-dimethylpropanethioate
  • VI243: 5′DMT 2′omethyl uridine (1 g, 1.8 mmol) and Ethylthioltetrazole 0.45M (7.1 ml) were added to a flame dried vial equipped with a stir bar. To the stirring solution was added 7 ml dichloromethane and the phosphorodiamidite VI253 (738.1 mg, 1.88 mmol) and the reaction was allowed to proceed for 3 h. The reaction concentrated to a viscous oil and Isolation was achieved by TEA pretreated silica chromatography. Fractions containing product were evaporated followed by transfer/filtration through a 0.45 um filter in acetonitrile and lyophilized from benzene overnight to ensure complete removal of water and residual triethylamine. 1.3 g of lyophilate was recovered at 85.4%.
  • Alternate post oligonucleotide synthesis routes to phosphotriesters:
  • Figure US20090093425A1-20090409-C00062
  • Methylphosphonates of the general structure listed above also provide access to oligonucleotides with charge neutral backbones that additionally provide nuclease resistance.
  • The examples set forth above are provided to give those of ordinary skill in the art a complete disclosure and description of how to make and use the embodiments of the apparatus, systems and methods of the disclosure, and are not intended to limit the scope of what the inventors regard as their disclosure. Modifications of the above-described modes for carrying out the disclosure that are obvious to persons of skill in the art are intended to be within the scope of the following claims. All patents and publications mentioned in the specification are indicative of the levels of skill of those skilled in the art to which the disclosure pertains. All references cited in this disclosure are incorporated by reference to the same extent as if each reference had been incorporated by reference in its entirety individually.
  • A number of embodiments of the disclosure have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the disclosure. Accordingly, other embodiments are within the scope of the following claims.

Claims (36)

1. A nucleic acid construct comprising:
a) an oligonucleotide or polynucleotide domain comprising a phosphodiester and/or phosphothioate protecting group at one or more positions that reduces the net anionic charge of the oligonucleotide or polynucleotide backbone; and
b) at least one transduction domain comprising a membrane transport function operably linked to the oligonucleotide or polynucleotide domain.
2. The nucleic acid construct of claim 1, wherein the at least one transduction domain is a cationically charged domain.
3. The nucleic acid construct of claim 1, wherein the at least one transduction domain is a protein transduction domain (PTD).
4. The nucleic acid construct of claim 1, wherein overall charge of the nucleic acid construct is neutral or positively charged relative to a non-protected oligonucleotide or polynucleotide.
5. The nucleic acid construct of claim 1, wherein the phosphodiester and/or phosphothioate protecting group has the general formula:

R—X— or Q-X—
wherein X is O, S or NR1, and R1 is H, methyl, ethyl, S-pivaloyl thioethanol, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
wherein R is selected from the group consisting of:

R2,
wherein R2 is alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic,
Figure US20090093425A1-20090409-C00063
wherein R3 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
wherein A1 and A2 are each independently one to seven atom chains, or substituted one to seven atom chains,
Figure US20090093425A1-20090409-C00064
wherein R4 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
wherein A3 and A4 are each independently one to seven atom chains, or substituted one to seven atom chains,
Figure US20090093425A1-20090409-C00065
wherein R5 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
wherein A5 is a one to seven atom chain, or substituted one to seven atom chain,
wherein X1 and X2 are each independently O, S or NR7, and R7 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
Figure US20090093425A1-20090409-C00066
wherein R6 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
wherein A6 and A6 are each independently one to seven atom chains, or substituted one to seven atom chains,
wherein X3 and X4 are each independently O, S or NR8, and R8 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
wherein Q is selected from the group consisting of:

Q1,
wherein Q1 is a basic group with a pKa greater than or equal to 10,
Figure US20090093425A1-20090409-C00067
wherein Q2 is a basic group with a pKa greater than or equal to 10,
wherein A8 and A9 are each independently one to seven atom chains, or substituted one to seven atom chains,
Figure US20090093425A1-20090409-C00068
wherein Q3 is a basic group with a pKa greater than or equal to 10,
wherein A10 and A11 are each independently one to seven atom chains, or substituted one to seven atom chains,
Figure US20090093425A1-20090409-C00069
wherein Q4 is a basic group with a pKa greater than or equal to 10,
wherein A12 is a one to seven atom chain, or substituted one to seven atom chain,
wherein X5 and X6 are each independently O, S or NR9, and R9 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
Figure US20090093425A1-20090409-C00070
wherein Q5 is a basic group with a pKa greater than or equal to 10,
wherein A13 and A14 are each independently one to seven atom chains, or substituted one to seven atom chains,
wherein X7 and X8 are each independently O, S or NR10, and R10 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic.
6. The nucleic acid construct of claim 1, wherein the phosphodiester and/or phosphothioate protecting group is selected from the group consisting of MeO—, EtO—, iPrO,
Figure US20090093425A1-20090409-C00071
7. The nucleic acid construct of claim 5, wherein the phosphodiester and/or phosphothioate protecting group comprises a structure selected from the group consisting of:
Figure US20090093425A1-20090409-C00072
8. The nucleic acid construct of claim 5, further comprising a basic substitution, guanidinium, amine, or urea group attached to the phosphodiester and/or phosphothioate protecting group via a linker to form an RNB protecting group.
9. The nucleic acid construct of claim 8, wherein the RNB protecting group comprises a structure selected from the group consisting of:
Figure US20090093425A1-20090409-C00073
10. The nucleic acid construct of claim 3, wherein the at least one protein transduction domain is selected from the group consisting of a polypeptide comprising a herpesviral VP22 domain; a polypeptide comprising a human immunodeficiency virus (HIV) TAT domain; a polypeptide comprising a homeodomain of an Antennapedia protein (Antp HD) domain; an N-terminal cationic prion protein domain; poly-Arg; and functional fragments thereof.
11. The nucleic acid construct of claim 1, wherein the oligonucleotide or polynucleotide domain comprises dsRNA or siRNA.
12. A pharmaceutical composition comprising the nucleic acid construct of claim 1 and a pharmaceutically acceptable carrier.
13. A nucleic acid construct comprising:
an oligonucleotide or polynucleotide comprising a phosphodiester and/or phosphothioate protecting group at one or more positions that reduces the net anionic charge of the oligonucleotide or polynucleotide backbone or provides a net cationic charge of the oligonucleotide or polynucleotide backbone.
14. The nucleic acid construct of claim 13, wherein the phosphodiester and/or phosphothioate protecting group has the general formula:

R—X— or Q-X—
wherein X is O, S or NR1, and R1 is H, methyl, ethyl, S-pivaloyl thioethanol, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
wherein R is selected from the group consisting of:

R2,
wherein R2 is alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic,
Figure US20090093425A1-20090409-C00074
wherein R3 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
wherein A1 and A2 are each independently one to seven atom chains, or substituted one to seven atom chains,
Figure US20090093425A1-20090409-C00075
wherein R4 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
wherein A3 and A4 are each independently one to seven atom chains, or substituted one to seven atom chains,
Figure US20090093425A1-20090409-C00076
wherein R5 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
wherein A5 is a one to seven atom chain, or substituted one to seven atom chain,
wherein X1 and X2 are each independently O, S or NR7, and R7 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
Figure US20090093425A1-20090409-C00077
wherein R6 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, substituted heterocyclic, halo, cyano, or nitro,
wherein A6 and A6 are each independently one to seven atom chains, or substituted one to seven atom chains,
wherein X3 and X4 are each independently O, S or NR8, and R8 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic;
wherein Q is selected from the group consisting of:

Q1,
wherein Q1 is a basic group with a pKa greater than or equal to 10,
Figure US20090093425A1-20090409-C00078
wherein Q2 is a basic group with a pKa greater than or equal to 10,
wherein A8 and A9 are each independently one to seven atom chains, or substituted one to seven atom chains,
Figure US20090093425A1-20090409-C00079
wherein Q3 is a basic group with a pKa greater than or equal to 10,
wherein A10 and A11 are each independently one to seven atom chains, or substituted one to seven atom chains,
Figure US20090093425A1-20090409-C00080
wherein Q4 is a basic group with a pKa greater than or equal to 10,
wherein A12 is a one to seven atom chain, or substituted one to seven atom chain,
wherein X5 and X6 are each independently O, S or NR9, and R9 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic, and
Figure US20090093425A1-20090409-C00081
wherein Q5 is a basic group with a pKa greater than or equal to 10,
wherein A13 and A14 are each independently one to seven atom chains, or substituted one to seven atom chains,
wherein X7 and X8 are each independently O, S or NR10, and R10 is H, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heterocyclic, or substituted heterocyclic.
15. The nucleic acid construct of claim 13, wherein the phosphodiester and/or phosphothioate protecting group is selected from the group consisting of MeO—, EtO—, iPrO,
Figure US20090093425A1-20090409-C00082
16. The nucleic acid construct of claim 14, wherein the phosphodiester and/or phosphothioate protecting group comprises a structure selected from the group consisting of:
Figure US20090093425A1-20090409-C00083
17. The nucleic acid construct of claim 14, comprising a basic substitution, guanidinium, amine, or urea group attached to the phosphodiester and/or phosphothioate protecting group via a linker to form an RNB protecting group.
18. The nucleic acid construct of claim 17, wherein the RNB protecting group comprises a structure selected from the group consisting of:
Figure US20090093425A1-20090409-C00084
19. The nucleic acid construct of claim 13, further comprising a targeting domain operably linked to the oligonucleotide or polynucleotide.
20. The nucleic acid construct of claim 19, wherein the targeting moiety is an antibody that binds to a cell surface protein.
21. The nucleic acid construct of claim 19, wherein the targeting domain comprises a receptor ligand for a cell surface receptor.
22. The nucleic acid construct of claim 13, further comprising at least one cationically charged polypeptide or peptide.
23. The nucleic acid construct of claim 22, wherein the cationically charged polypeptide or peptide comprises a protein transduction domain.
24. A pharmaceutical composition comprising the nucleic acid construct of claim 13, and a pharmaceutically acceptable carrier.
25. A method comprising:
linking one or more protein transduction domain to a nucleic acid construct of claim 13.
26. The method of claim 25, wherein the one or more protein transduction domains comprise 2-5 protein transduction domains.
27. The method of claim 26, wherein the one or more protein transduction domains comprise three protein transduction domains.
28. A method of generating a nucleic acid construct comprising:
substantially purifying a protein transduction domain;
synthesizing an oligonucleotide;
charge neutralizing the anionic charge on the oligonucleotide with a phosphotriester group; and
linking the oligonucleotide to one or more protein transduction domains.
29. A method of transfecting a cell, comprising:
contacting the cell with a nucleic acid construct of claim 1 or 13.
30. The method of claim 29, wherein the contacting is in vivo.
31. The method of claim 30, wherein the nucleic acid construct comprises an antisense molecule.
32. A method of treating a disease or disorder comprising administering a nucleic acid construct of claim 1 or 13 to a subject, wherein the oligonucleotide or polynucleotide comprises a therapeutic or diagnostic molecule.
33. The nucleic acid construct of claim 5 or 14, wherein R1 is a methyl, ethyl or S-pivaloyl thioethanol.
34. The nucleic acid construct of claim 33, wherein the phosphotriester and/or phosphothioate protecting group present on a nucleotide comprising a 2′-W, wherein W is H, F, O-Me, or O-Alkyl.
35. The nucleic acid construct of claim 10, wherein the at least one transduction domain comprises two or more transduction domains.
36. The nucleic acid of claim 1 or 35, wherein the at least one transduction domain is reversibly or irreversibly conjugated to the 5′, 3′, or 5′ and 3′ end of the oligonucleotide or polynucleotide domain.
US11/776,317 2006-07-12 2007-07-11 Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups Abandoned US20090093425A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/776,317 US20090093425A1 (en) 2006-07-12 2007-07-11 Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US83057206P 2006-07-12 2006-07-12
US11/776,317 US20090093425A1 (en) 2006-07-12 2007-07-11 Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups

Publications (1)

Publication Number Publication Date
US20090093425A1 true US20090093425A1 (en) 2009-04-09

Family

ID=38923923

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/776,317 Abandoned US20090093425A1 (en) 2006-07-12 2007-07-11 Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups

Country Status (7)

Country Link
US (1) US20090093425A1 (en)
EP (1) EP2046964B1 (en)
JP (2) JP6125741B2 (en)
CN (1) CN101506368B (en)
AU (1) AU2007272906B2 (en)
CA (1) CA2659103C (en)
WO (1) WO2008008476A2 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090093026A1 (en) * 2006-02-10 2009-04-09 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF siRNA BY dsRNA BINDING DOMAIN FUSIONS TO PTD/CPPS
WO2011109677A2 (en) * 2010-03-04 2011-09-09 University Of Louisville Research Foundation, Inc. Methods of increasing macropinocytosis in cancer cells
US20140031413A1 (en) * 2011-04-05 2014-01-30 The Regents Of The University Of California Method and compositions comprising small rna agonist and antagonists to modulate inflammation
WO2014043289A2 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to ddit4 and methods of use thereof
WO2014043292A1 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to p53 and methods of use thereof
US8691971B2 (en) 2008-09-23 2014-04-08 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating RNA interference
WO2014145254A2 (en) 2013-03-15 2014-09-18 Sutter West Bay Hospitals Falz for use as a target for therapies to treat cancer
WO2015015496A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Sphingolipid-polyalkylamine-oligonucleotide compounds
WO2015015498A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Methods of use of sphingolipid polyalkylamine oligonucleotide compounds
US9236276B2 (en) 2011-12-01 2016-01-12 Denso Corporation Semiconductor device and method for manufacturing the same
US9394333B2 (en) 2008-12-02 2016-07-19 Wave Life Sciences Japan Method for the synthesis of phosphorus atom modified nucleic acids
US9598458B2 (en) 2012-07-13 2017-03-21 Wave Life Sciences Japan, Inc. Asymmetric auxiliary group
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US9617547B2 (en) 2012-07-13 2017-04-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US10022454B2 (en) 2008-09-23 2018-07-17 Liposciences, Llc Functionalized phosphorodiamites for therapeutic oligonucleotide synthesis
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US11597744B2 (en) 2017-06-30 2023-03-07 Sirius Therapeutics, Inc. Chiral phosphoramidite auxiliaries and methods of their use

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010129853A2 (en) * 2009-05-07 2010-11-11 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF NUCLEIC ACIDS USING MODIFIED dsRNA BINDING DOMAINS
CN102459302A (en) * 2009-06-01 2012-05-16 加利福尼亚大学董事会 Nucleic acid delivery compositions and methods of use thereof
EP4089169A1 (en) 2009-10-12 2022-11-16 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
EP3919620A1 (en) 2012-05-02 2021-12-08 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions
AU2013306006B2 (en) 2012-08-20 2017-07-06 The Regents Of The University Of California Polynucleotides having bioreversible groups
WO2014088920A1 (en) * 2012-12-06 2014-06-12 Merck Sharp & Dohme Corp. Disulfide masked prodrug compositions and methods
RU2015126795A (en) * 2012-12-06 2017-01-12 Мерк Шарп И Доум Корп. MINUS NUCLEOSIDE KINASE COMPOSITIONS AND METHODS FOR PRODUCING AND USING THEM
KR20210123433A (en) * 2013-08-29 2021-10-13 시티 오브 호프 Cell penetrating conjugates and methods of use thereof
GB201410507D0 (en) * 2014-06-12 2014-07-30 Univ Bath Drug delivery enhancement agents
AU2017312116A1 (en) * 2016-08-17 2019-03-07 Solstice Biologics, Ltd. Polynucleotide constructs
US11236334B2 (en) 2017-08-22 2022-02-01 National University Corporation Nagoya University Modified polynucleotide

Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US5670617A (en) * 1989-12-21 1997-09-23 Biogen Inc Nucleic acid conjugates of tat-derived transport polypeptides
US5733523A (en) * 1990-12-10 1998-03-31 Akzo Nobel N.V. Targeted delivery of a therapeutic entity using complementary oligonucleotides
US5955591A (en) * 1993-05-12 1999-09-21 Imbach; Jean-Louis Phosphotriester oligonucleotides, amidites and method of preparation
US6022735A (en) * 1991-09-30 2000-02-08 Boehringer Ingelheim International Gmbh Composition for introducing nucleic acid complexes into higher eucaryotic cells
US6221355B1 (en) * 1997-12-10 2001-04-24 Washington University Anti-pathogen system and methods of use thereof
US6316003B1 (en) * 1989-12-21 2001-11-13 Whitehead Institute For Biomedical Research Tat-derived transport polypeptides
US6376248B1 (en) * 1997-03-14 2002-04-23 Life Technologies, Inc. Peptide-enhanced transfections
US6468986B1 (en) * 1996-08-13 2002-10-22 Chiron Corporation Compositions and methods for polynucleotide delivery
US20030143732A1 (en) * 2001-04-05 2003-07-31 Kathy Fosnaugh RNA interference mediated inhibition of adenosine A1 receptor (ADORA1) gene expression using short interfering RNA
US20040204377A1 (en) * 2002-11-26 2004-10-14 University Of Massachusetts Delivery of siRNAs
US6841535B2 (en) * 2000-07-31 2005-01-11 Active Motif Peptide-mediated transfection agents and methods of use
US20050042603A1 (en) * 2003-07-01 2005-02-24 Allele Biotechnology & Pharmaceuticals, Inc. Compositions and methods for peptide-assisted transfection
US6903077B1 (en) * 1999-01-04 2005-06-07 University Of Vermont And State Agricultural College Methods and products for delivering nucleic acids
US20050147993A1 (en) * 2003-10-24 2005-07-07 Shaharyar Khan Methods and compositions for delivering polynucleotides
US20050239687A1 (en) * 2004-04-26 2005-10-27 Centre National De La Recherche Scientifique Methods and compositions for delivering siRNA into mammalian cells
US20050260756A1 (en) * 2003-01-28 2005-11-24 Troy Carol M Complex for facilitating delivery of dsRNA into a cell and uses thereof
US20060030003A1 (en) * 2004-05-12 2006-02-09 Simon Michael R Composition and method for introduction of RNA interference sequences into targeted cells and tissues
US20060035815A1 (en) * 2004-05-04 2006-02-16 Nastech Pharmaceutical Company Inc. Pharmaceutical compositions for delivery of ribonucleic acid to a cell
US20060040882A1 (en) * 2004-05-04 2006-02-23 Lishan Chen Compostions and methods for enhancing delivery of nucleic acids into cells and for modifying expression of target genes in cells
US7084248B2 (en) * 2004-07-14 2006-08-01 Gene Tools, Llc Peptide composition and method for delivering substances into the cytosol of cells
US20060178297A1 (en) * 2003-01-28 2006-08-10 Troy Carol M Systems and methods for silencing expression of a gene in a cell and uses thereof
US20060182736A1 (en) * 2003-06-10 2006-08-17 Kim Jin-Soo Transducible dna-binding proteins
US20060205665A1 (en) * 2000-10-13 2006-09-14 Christophe Bonny Intracellular delivery of biological effectors
US20060222657A1 (en) * 2003-06-20 2006-10-05 Dowdy Steven F Polypeptide transduction and fusogenic peptides
US7166692B2 (en) * 2003-03-04 2007-01-23 Canbrex Bio Science Walkersville, Inc. Intracellular delivery of small molecules, proteins, and nucleic acids
US20080027025A1 (en) * 2004-08-27 2008-01-31 The Regents Of The University Of California Protection From And Treatment Of Prion Protein Infection
US20080153737A1 (en) * 2004-08-16 2008-06-26 The Cbr Institute For Biomedical Research, Inc. Method of Delivering Rna Interference and Uses Thereof
US7491805B2 (en) * 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US20090093026A1 (en) * 2006-02-10 2009-04-09 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF siRNA BY dsRNA BINDING DOMAIN FUSIONS TO PTD/CPPS
US20090098049A1 (en) * 2004-09-07 2009-04-16 The Regents Of The University Of California Targeting transducible molecules to specific cell types
US20120142763A1 (en) * 2009-06-01 2012-06-07 The Regents Of The University Of California Nucleic acid delivery compositions and methods of use thereof

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991014696A1 (en) * 1990-03-29 1991-10-03 Gilead Sciences, Inc. Oligonucleotide-transport agent disulfide conjugates
US6017700A (en) * 1995-08-04 2000-01-25 Bayer Corporation Cationic oligonucleotides, and related methods of synthesis and use
US6531590B1 (en) * 1998-04-24 2003-03-11 Isis Pharmaceuticals, Inc. Processes for the synthesis of oligonucleotide compounds
JP2007536253A (en) * 2004-05-04 2007-12-13 ナステック・ファーマシューティカル・カンパニー・インコーポレーテッド Compositions and methods for enhancing delivery of nucleic acids into cells and modifying expression of target genes in cells
US20050288246A1 (en) * 2004-05-24 2005-12-29 Iversen Patrick L Peptide conjugated, inosine-substituted antisense oligomer compound and method
WO2006007721A1 (en) * 2004-07-21 2006-01-26 Xiao Huang Hybrid wicking materials for use in high performance heat pipes
CN101421301A (en) * 2006-02-10 2009-04-29 加利福尼亚大学董事会 Transducible delivery of sirna by dsrna binding domain fusions to ptd/cpps

Patent Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US6316003B1 (en) * 1989-12-21 2001-11-13 Whitehead Institute For Biomedical Research Tat-derived transport polypeptides
US5670617A (en) * 1989-12-21 1997-09-23 Biogen Inc Nucleic acid conjugates of tat-derived transport polypeptides
US5733523A (en) * 1990-12-10 1998-03-31 Akzo Nobel N.V. Targeted delivery of a therapeutic entity using complementary oligonucleotides
US6022735A (en) * 1991-09-30 2000-02-08 Boehringer Ingelheim International Gmbh Composition for introducing nucleic acid complexes into higher eucaryotic cells
US6077663A (en) * 1991-09-30 2000-06-20 Boehringer Ingelheim International Gmbh Composition for introducing nucleic acid complexes into higher eucaryotic cells
US5955591A (en) * 1993-05-12 1999-09-21 Imbach; Jean-Louis Phosphotriester oligonucleotides, amidites and method of preparation
US6124445A (en) * 1994-11-23 2000-09-26 Isis Pharmaceuticals, Inc. Phosphotriester oligonucleotides, amidities and method of preparation
US6468986B1 (en) * 1996-08-13 2002-10-22 Chiron Corporation Compositions and methods for polynucleotide delivery
US6376248B1 (en) * 1997-03-14 2002-04-23 Life Technologies, Inc. Peptide-enhanced transfections
US6645501B2 (en) * 1997-12-10 2003-11-11 Washington University Anti-pathogen system and methods of use thereof
US6221355B1 (en) * 1997-12-10 2001-04-24 Washington University Anti-pathogen system and methods of use thereof
US6903077B1 (en) * 1999-01-04 2005-06-07 University Of Vermont And State Agricultural College Methods and products for delivering nucleic acids
US6841535B2 (en) * 2000-07-31 2005-01-11 Active Motif Peptide-mediated transfection agents and methods of use
US20060205665A1 (en) * 2000-10-13 2006-09-14 Christophe Bonny Intracellular delivery of biological effectors
US20030143732A1 (en) * 2001-04-05 2003-07-31 Kathy Fosnaugh RNA interference mediated inhibition of adenosine A1 receptor (ADORA1) gene expression using short interfering RNA
US7491805B2 (en) * 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US20040204377A1 (en) * 2002-11-26 2004-10-14 University Of Massachusetts Delivery of siRNAs
US20050260756A1 (en) * 2003-01-28 2005-11-24 Troy Carol M Complex for facilitating delivery of dsRNA into a cell and uses thereof
US20060178297A1 (en) * 2003-01-28 2006-08-10 Troy Carol M Systems and methods for silencing expression of a gene in a cell and uses thereof
US7420031B2 (en) * 2003-03-04 2008-09-02 Lonza Walkersville, Inc. Intracellular delivery of small molecules, proteins, and nucleic acids
US7166692B2 (en) * 2003-03-04 2007-01-23 Canbrex Bio Science Walkersville, Inc. Intracellular delivery of small molecules, proteins, and nucleic acids
US20060182736A1 (en) * 2003-06-10 2006-08-17 Kim Jin-Soo Transducible dna-binding proteins
US20060222657A1 (en) * 2003-06-20 2006-10-05 Dowdy Steven F Polypeptide transduction and fusogenic peptides
US20050042603A1 (en) * 2003-07-01 2005-02-24 Allele Biotechnology & Pharmaceuticals, Inc. Compositions and methods for peptide-assisted transfection
US20050147993A1 (en) * 2003-10-24 2005-07-07 Shaharyar Khan Methods and compositions for delivering polynucleotides
US7514530B2 (en) * 2004-04-26 2009-04-07 Centre National De La Recherche Scientifique Peptide carrier for delivering siRNA into mammalian cells
US20050239687A1 (en) * 2004-04-26 2005-10-27 Centre National De La Recherche Scientifique Methods and compositions for delivering siRNA into mammalian cells
US20060040882A1 (en) * 2004-05-04 2006-02-23 Lishan Chen Compostions and methods for enhancing delivery of nucleic acids into cells and for modifying expression of target genes in cells
US20060035815A1 (en) * 2004-05-04 2006-02-16 Nastech Pharmaceutical Company Inc. Pharmaceutical compositions for delivery of ribonucleic acid to a cell
US20060030003A1 (en) * 2004-05-12 2006-02-09 Simon Michael R Composition and method for introduction of RNA interference sequences into targeted cells and tissues
US7084248B2 (en) * 2004-07-14 2006-08-01 Gene Tools, Llc Peptide composition and method for delivering substances into the cytosol of cells
US20080153737A1 (en) * 2004-08-16 2008-06-26 The Cbr Institute For Biomedical Research, Inc. Method of Delivering Rna Interference and Uses Thereof
US20080027025A1 (en) * 2004-08-27 2008-01-31 The Regents Of The University Of California Protection From And Treatment Of Prion Protein Infection
US20090098049A1 (en) * 2004-09-07 2009-04-16 The Regents Of The University Of California Targeting transducible molecules to specific cell types
US20090093026A1 (en) * 2006-02-10 2009-04-09 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF siRNA BY dsRNA BINDING DOMAIN FUSIONS TO PTD/CPPS
US20120142763A1 (en) * 2009-06-01 2012-06-07 The Regents Of The University Of California Nucleic acid delivery compositions and methods of use thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Gillet et al (Nucl. Acids Res. 33(6): 1961-1969, 2005) *
Hayakawa (Bull. Chem. Soc. Jpn., 74, No. 9 (2001)) *
Parrish et al., (Molecular Cell, 6, 1977-1087, 2000) *
Spinelli et al (Eur. J. Org. Chem. 2002, 49256) *

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8273867B2 (en) 2006-02-10 2012-09-25 The Regents Of The University Of California Transducible delivery of siRNA by dsRNA binding domain fusions to PTD/CPPS
US20090093026A1 (en) * 2006-02-10 2009-04-09 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF siRNA BY dsRNA BINDING DOMAIN FUSIONS TO PTD/CPPS
US8691971B2 (en) 2008-09-23 2014-04-08 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating RNA interference
US10022454B2 (en) 2008-09-23 2018-07-17 Liposciences, Llc Functionalized phosphorodiamites for therapeutic oligonucleotide synthesis
US9394333B2 (en) 2008-12-02 2016-07-19 Wave Life Sciences Japan Method for the synthesis of phosphorus atom modified nucleic acids
US9695211B2 (en) 2008-12-02 2017-07-04 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
US10329318B2 (en) 2008-12-02 2019-06-25 Wave Life Sciences Ltd. Method for the synthesis of phosphorus atom modified nucleic acids
US10307434B2 (en) 2009-07-06 2019-06-04 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
WO2011109677A2 (en) * 2010-03-04 2011-09-09 University Of Louisville Research Foundation, Inc. Methods of increasing macropinocytosis in cancer cells
WO2011109677A3 (en) * 2010-03-04 2012-02-23 University Of Louisville Research Foundation, Inc. Methods of increasing macropinocytosis in cancer cells
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US20140031413A1 (en) * 2011-04-05 2014-01-30 The Regents Of The University Of California Method and compositions comprising small rna agonist and antagonists to modulate inflammation
US9303258B2 (en) * 2011-04-05 2016-04-05 The Regents Of The University Of California Method and compositions comprising small RNA agonist and antagonists to modulate inflammation
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US10280192B2 (en) 2011-07-19 2019-05-07 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US9236276B2 (en) 2011-12-01 2016-01-12 Denso Corporation Semiconductor device and method for manufacturing the same
US10167309B2 (en) 2012-07-13 2019-01-01 Wave Life Sciences Ltd. Asymmetric auxiliary group
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US9598458B2 (en) 2012-07-13 2017-03-21 Wave Life Sciences Japan, Inc. Asymmetric auxiliary group
US9617547B2 (en) 2012-07-13 2017-04-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
US10590413B2 (en) 2012-07-13 2020-03-17 Wave Life Sciences Ltd. Chiral control
WO2014043292A1 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to p53 and methods of use thereof
WO2014043289A2 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to ddit4 and methods of use thereof
WO2014145254A2 (en) 2013-03-15 2014-09-18 Sutter West Bay Hospitals Falz for use as a target for therapies to treat cancer
WO2015015498A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Methods of use of sphingolipid polyalkylamine oligonucleotide compounds
WO2015015496A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Sphingolipid-polyalkylamine-oligonucleotide compounds
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US11597744B2 (en) 2017-06-30 2023-03-07 Sirius Therapeutics, Inc. Chiral phosphoramidite auxiliaries and methods of their use

Also Published As

Publication number Publication date
WO2008008476A2 (en) 2008-01-17
JP2015062421A (en) 2015-04-09
AU2007272906A1 (en) 2008-01-17
EP2046964A4 (en) 2014-05-14
CA2659103A1 (en) 2008-01-17
CN101506368A (en) 2009-08-12
WO2008008476A3 (en) 2008-07-24
AU2007272906B2 (en) 2013-01-31
CA2659103C (en) 2019-05-21
JP6125741B2 (en) 2017-05-17
JP2009543783A (en) 2009-12-10
EP2046964A2 (en) 2009-04-15
CN101506368B (en) 2017-02-08
EP2046964B1 (en) 2018-07-04

Similar Documents

Publication Publication Date Title
US20090093425A1 (en) Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups
US20120142763A1 (en) Nucleic acid delivery compositions and methods of use thereof
US20200392498A1 (en) Polynucleotide constructs having bioreversible and non-bioreversible groups
US8691971B2 (en) Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating RNA interference
US20180303864A1 (en) Polynucleotides having bioreversible groups
JP6048982B2 (en) Artificial nucleosides and oligonucleotides with guanidine bridges
US20160257961A1 (en) Polynucleotide constructs having disulfide groups
CA3120580A1 (en) Rnai agents for inhibiting expression of hif-2 alpha (epas1), compositions thereof, and methods of use
US10022454B2 (en) Functionalized phosphorodiamites for therapeutic oligonucleotide synthesis
WO2020044349A1 (en) Compounds, cojugates and compositions for use in the methods for trans-membrane delivery of molecules
US20230002767A1 (en) RNAi Agents for Inhibiting Expression of Mucin 5AC (MUC5AC), Compositions Thereof, and Methods of Use
CN114846142A (en) RNAi agents for inhibiting expression of beta-ENaC, compositions thereof, and methods of use
CN117440817A (en) RNAi agents for inhibiting expression of advanced glycation end product receptors, compositions and methods of use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENTS OF THE UNIVERSITY OF CALIFORNIA, THE, CALI

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DOWDY, STEVEN F.;PETERSEN, SCOTT G.;MEADE, BRYAN R.;REEL/FRAME:019559/0415

Effective date: 20070710

Owner name: HOWARD HUGHES MEDICAL INSTITUTE, THE, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DOWDY, STEVEN F.;PETERSEN, SCOTT G.;REEL/FRAME:019559/0460;SIGNING DATES FROM 20060823 TO 20060913

STCV Information on status: appeal procedure

Free format text: ON APPEAL -- AWAITING DECISION BY THE BOARD OF APPEALS

STCV Information on status: appeal procedure

Free format text: BOARD OF APPEALS DECISION RENDERED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION